1
|
Sivanantharajah L, Mudher A, Shepherd D. Examining the vulnerability of adult neuron subtypes to tau-mediated toxicity in Drosophila. Transl Psychiatry 2025; 15:127. [PMID: 40188067 PMCID: PMC11972385 DOI: 10.1038/s41398-025-03342-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 02/26/2025] [Accepted: 03/20/2025] [Indexed: 04/07/2025] Open
Abstract
Selective vulnerability of nerve cells is a feature of neurodegenerative disease. To date, animal models have been limited to examining pathogenic protein expression in broad or heterogeneous neuronal populations. Consequently, noted pathological hallmarks represent an average of disease phenotypes over multiple neuron types, rather than exact measures of individual responses. Here we targeted gene expression to small, precisely defined and homogenous neuronal populations in the Drosophila melanogaster central nervous system (CNS), allowing dissection of selective vulnerability of single types of neurons with single-neuron resolution. Using cellular degeneration as a readout for vulnerability, we found while all neurons were affected by tau some neuron types were more affected (vulnerable) than others (resilient). The tau-mediated pathogenic effects fell on a spectrum, demonstrating that neurons in the fly CNS are differentially vulnerable to tau pathology. Mechanistically, total tau levels did not correlate with vulnerability; rather, the best correlatives of degeneration were significant age-dependent increases in phospho-tau levels in the same neuron type, and tau mislocalisation into dendrites. Lastly, we found that tau phosphorylation in vulnerable neuron types correlated with downstream vesicular and mitochondrial trafficking defects. However, all vulnerable neuron types did not show the same pattern, suggesting multiple paths to degeneration. Beyond highlighting the heterogeneity of neuronal responses to tau in determining vulnerability, this work provides a new, high-resolution, tractable model for studying the age-dependent effects of tau, or any pathogenic protein, on postmitotic neurons with sub-cellular resolution.
Collapse
Affiliation(s)
| | - Amrit Mudher
- Faculty of Natural and Environmental Sciences, University of Southampton, Southampton, UK
| | - David Shepherd
- Faculty of Natural and Environmental Sciences, University of Southampton, Southampton, UK
| |
Collapse
|
2
|
Arora R, Deshmukh R. Embelin Mitigates Amyloid-β-Induced Neurotoxicity and Cognitive Impairment in Rats: Potential Therapeutic Implications for Alzheimer's Disease. Mol Neurobiol 2025; 62:1577-1590. [PMID: 39008170 DOI: 10.1007/s12035-024-04308-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 06/13/2024] [Indexed: 07/16/2024]
Abstract
Alzheimer's disease (AD) is a significant form of dementia. Embelin (EMB) is a natural compound with varied actions that could help prevent AD pathology. Herein, we have investigated the neuroprotective potential of EMB against Aβ1-42-induced neurotoxicity in rats. In this experiment, Alzheimer-like dementia was induced in rats by infusing Aβ1-42 oligomers directly into the brain's ventricles. Subsequently, the Aβ1-42-intoxicated rats received treatment with varying doses of EMB (2.5, 5, and 10 mg/kg, administered intraperitoneally) over 2 weeks. The spatial and non-spatial memory of animals was assessed at different time intervals, and various biochemical, neurochemical, and neuroinflammatory parameters in the hippocampal brain tissue of the rats were analyzed. Infusion of Aβ1-42 in rat brain caused cognitive impairment and was accompanied by increased acetylcholinesterase activity, oxidative stress, and elevated levels of pro-inflammatory cytokines (such as TNF-α, IL-1β, and IL-6) in the hippocampal tissue. Moreover, a significant decline in the levels of monoamines and an imbalance of GABA and glutamate levels were also observed. EMB treatment significantly mitigated Aβ1-42-induced cognitive deficit and other biochemical changes, including Aβ levels. The EMB-treated rats showed improved learning and consolidation of memory. EMB also attenuated Aβ-induced oxidative stress and neuroinflammation and restored the levels of monoamines and the balance between GABA and glutamate. The observed cognitive benefits following EMB treatment in Aβ1-42-infused rats may be attributed to its antioxidant and anti-inflammatory properties and ability to restore hippocampal neurochemistry and Aβ levels. The above findings indicate the therapeutic potential of EMB in neurodegenerative pathologies associated with cognitive decline, such as Alzheimer's disease.
Collapse
Affiliation(s)
- Rimpi Arora
- Research Scholar, IKGPTU, Jalandhar, Punjab, India;, ISF College of Pharmacy, Moga, Pb, 142001, India
| | - Rahul Deshmukh
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda, Pb, 151001, India.
| |
Collapse
|
3
|
Sawarkar P, Yadav N, Sawarkar G. Management of Spinocerebellar Ataxia through Ayurveda-A Case Report. JOURNAL OF PHARMACY AND BIOALLIED SCIENCES 2024; 16:S4158-S4160. [PMID: 39926966 PMCID: PMC11805052 DOI: 10.4103/jpbs.jpbs_957_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 08/19/2024] [Accepted: 09/07/2024] [Indexed: 02/11/2025] Open
Abstract
Spinocerebellar ataxias (SCA) are neurodegenerative disorders marked by progressive cerebellar ataxia, impacting balance, motor coordination, and ocular function due to degeneration. This study evaluated Shodhana Chikitsa and Shaman Chikitsa (purification and symptomatic treatments) for SCA. A 27-year-old man with SCA, showing symptoms like swaying, poor balance, and slurred speech, was referred to the Panchakarma OPD at Ayurveda Hospital, Salod, Wardha. He underwent a comprehensive regimen, including full-body massage with herbal poultices (Patrapottali and Shashtishali Swedana), nasal instillation with medicated oil (Marsha Nasya), and a medicated enema with decoctions and oil, along with oral medications, such as Yogaraja Guggulu, Rasnasaptaka Qwath, Cap Neuron Plus, Agnitundi Vati, and Mahavatavidhwansa Rasa, for two months. Panchtikta Ksheer Vasti (medicated milk enema) was given for eight days. Significant improvements in symptoms and the Scale for Assessment and Rating of Ataxia (SARA) score were observed, suggesting that Ayurvedic treatments may be effective for SCA, warranting further research.
Collapse
Affiliation(s)
- Punam Sawarkar
- Department of Panchakarma, Mahatma Gandhi Ayurved College Hospital and Research Centre, Datta Meghe Institute of Higher Education and Research, Maharashtra, India
| | - Natasha Yadav
- Department of Dravyaguna Vigyan, State Government Ayurveda Yoga and Naturopathy College, Jaipur, Rajasthan, India
| | - Gaurav Sawarkar
- Department of Rachana Sharir, Mahatma Gandhi Ayurved College Hospital, Research Centre, Wardha, Maharashtra, India
- Department of Panchakarma, Datta Meghe Institute of Higher Education and Research, Wardha, Maharashtra, India
| |
Collapse
|
4
|
Slota JA, Lamoureux L, Frost KL, Sajesh BV, Booth SA. Single-cell transcriptomics unveils molecular signatures of neuronal vulnerability in a mouse model of prion disease that overlap with Alzheimer's disease. Nat Commun 2024; 15:10174. [PMID: 39580485 PMCID: PMC11585576 DOI: 10.1038/s41467-024-54579-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 11/14/2024] [Indexed: 11/25/2024] Open
Abstract
Understanding why certain neurons are more sensitive to dysfunction and death caused by misfolded proteins could provide therapeutically relevant insights into neurodegenerative disorders. Here, we harnessed single-cell transcriptomics to examine live neurons isolated from prion-infected female mice, aiming to identify and characterize prion-vulnerable neuronal subsets. Our analysis revealed distinct transcriptional responses across neuronal subsets, with a consistent pathway-level depletion of synaptic gene expression in damage-vulnerable neurons. By scoring neuronal damage based on the magnitude of depleted synaptic gene expression, we identified a diverse spectrum of prion-vulnerable glutamatergic, GABAergic, and medium spiny neurons. Comparison between prion-vulnerable and resistant neurons highlighted baseline gene expression differences that could influence neuronal vulnerability. For instance, the neuroprotective cold-shock protein Rbm3 exhibited higher baseline gene expression in prion-resistant neurons and was robustly upregulated across diverse neuronal classes upon prion infection. We also identified vulnerability-correlated transcripts that overlapped between prion and Alzheimer's disease. Our findings not only demonstrate the potential of single-cell transcriptomics to identify damage-vulnerable neurons, but also provide molecular insights into neuronal vulnerability and highlight commonalties across neurodegenerative disorders.
Collapse
Affiliation(s)
- Jessy A Slota
- Mycobacteriology, Vector-Borne and Prion Diseases Division, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
- Department of Medical Microbiology and Infectious Diseases, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Lise Lamoureux
- Mycobacteriology, Vector-Borne and Prion Diseases Division, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - Kathy L Frost
- Mycobacteriology, Vector-Borne and Prion Diseases Division, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - Babu V Sajesh
- Mycobacteriology, Vector-Borne and Prion Diseases Division, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - Stephanie A Booth
- Mycobacteriology, Vector-Borne and Prion Diseases Division, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada.
- Department of Medical Microbiology and Infectious Diseases, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
5
|
Groveman BR, Race B, Hughson AG, Haigh CL. Sodium hypochlorite inactivation of human CJD prions. PLoS One 2024; 19:e0312837. [PMID: 39509453 PMCID: PMC11542847 DOI: 10.1371/journal.pone.0312837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 10/15/2024] [Indexed: 11/15/2024] Open
Abstract
Prion diseases are transmissible, fatal neurologic diseases of mammals caused by the accumulation of mis-folded, disease associated prion protein (PrPd). Creutzfeldt-Jakob Disease (CJD) is the most common human prion disease and can occur by sporadic onset (sCJD) (~85% of CJD cases), genetic mutations in the prion protein gene (10-15%) or iatrogenic transmission (rare). PrPd is difficult to inactivate and many methods to reduce prion infectivity are dangerous, caustic, expensive, or impractical. Identifying viable and safe methods for decontamination of CJD exposed materials is critically important for medical facilities and research institutions. Previous research has shown that concentrated sodium hypochlorite (bleach) was effective at inactivation of CJD prions derived from brains of mice or guinea pigs. Unfortunately, human prions adapted to rodents may mis-fold differently than in humans, and the rodent adapted prions may not have the same resistance or susceptibility to inactivation present in bona fide CJD prions. To confirm that bleach was efficacious against human sourced CJD prions, we exposed different subtypes of sCJD-infected human brain homogenates to different concentrations of bleach for increasing exposure times. Initial and residual prion seeding activity following inactivation were measured using Real-Time Quaking Induced Conversion. In addition, we tested how passage of human sCJD into either transgenic mice that expressed human prion protein, or transmission of CJD to human cerebral organoids (CO), two common laboratory practices, may affect CJD prions' susceptibility to bleach inactivation. Our results show that bleach is effective against human sourced sCJD prions, and both treatment time and concentration of bleach were important factors for CJD inactivation. CJD derived from human brains, transgenic mouse brains or CO were all susceptible to inactivation with as low as a 10 percent bleach solution with a 30-minute exposure time or a 50 percent bleach solution with as little as a 1-minute exposure time.
Collapse
Affiliation(s)
- Bradley R. Groveman
- The Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Brent Race
- The Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Andrew G. Hughson
- The Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Cathryn L. Haigh
- The Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| |
Collapse
|
6
|
Kelser BM, Teichner EM, Subtirelu RC, Hoss KN. A review of proposed mechanisms for neurodegenerative disease. Front Aging Neurosci 2024; 16:1370580. [PMID: 39439710 PMCID: PMC11493710 DOI: 10.3389/fnagi.2024.1370580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 09/24/2024] [Indexed: 10/25/2024] Open
Abstract
Neurodegenerative diseases, such as Alzheimer's, Parkinson's, and amyotrophic lateral sclerosis (ALS) affect millions and present significant challenges in healthcare and treatment costs. The debate in the field pivots around two hypotheses: synaptic spread and selective vulnerability. Pioneers like Virginia Lee and John Trojanowski have been instrumental in identifying key proteins (tau, alpha-synuclein, TDP-43) central to these diseases. The synaptic spread hypothesis suggests a cell-to-cell propagation of pathogenic proteins across neuronal synapses, influencing disease progression, with studies highlighting the role of proteins like alpha-synuclein and amyloid-beta in this process. In contrast, the selective vulnerability hypothesis proposes inherent susceptibility of certain neurons to degeneration due to factors like metabolic stress, leading to protein aggregation. Recent advancements in neuroimaging, especially PET/MRI hybrid imaging, offer new insights into these mechanisms. While both hypotheses offer substantial evidence, their relative contributions to neurodegenerative processes remain to be fully elucidated. This uncertainty underscores the necessity for continued research, with a focus on these hypotheses, to develop effective treatments for these devastating diseases.
Collapse
Affiliation(s)
- Benjamin M. Kelser
- School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Eric M. Teichner
- Sidney Kimmel Medical College (SKMC), Philadelphia, PA, United States
| | - Robert C. Subtirelu
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Kevin N. Hoss
- School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
7
|
Moldovean-Cioroianu NS. Reviewing the Structure-Function Paradigm in Polyglutamine Disorders: A Synergistic Perspective on Theoretical and Experimental Approaches. Int J Mol Sci 2024; 25:6789. [PMID: 38928495 PMCID: PMC11204371 DOI: 10.3390/ijms25126789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 06/13/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
Polyglutamine (polyQ) disorders are a group of neurodegenerative diseases characterized by the excessive expansion of CAG (cytosine, adenine, guanine) repeats within host proteins. The quest to unravel the complex diseases mechanism has led researchers to adopt both theoretical and experimental methods, each offering unique insights into the underlying pathogenesis. This review emphasizes the significance of combining multiple approaches in the study of polyQ disorders, focusing on the structure-function correlations and the relevance of polyQ-related protein dynamics in neurodegeneration. By integrating computational/theoretical predictions with experimental observations, one can establish robust structure-function correlations, aiding in the identification of key molecular targets for therapeutic interventions. PolyQ proteins' dynamics, influenced by their length and interactions with other molecular partners, play a pivotal role in the polyQ-related pathogenic cascade. Moreover, conformational dynamics of polyQ proteins can trigger aggregation, leading to toxic assembles that hinder proper cellular homeostasis. Understanding these intricacies offers new avenues for therapeutic strategies by fine-tuning polyQ kinetics, in order to prevent and control disease progression. Last but not least, this review highlights the importance of integrating multidisciplinary efforts to advancing research in this field, bringing us closer to the ultimate goal of finding effective treatments against polyQ disorders.
Collapse
Affiliation(s)
- Nastasia Sanda Moldovean-Cioroianu
- Institute of Materials Science, Bioinspired Materials and Biosensor Technologies, Kiel University, Kaiserstraße 2, 24143 Kiel, Germany;
- Faculty of Physics, Babeș-Bolyai University, Kogălniceanu 1, RO-400084 Cluj-Napoca, Romania
| |
Collapse
|
8
|
Pak V, Adewale Q, Bzdok D, Dadar M, Zeighami Y, Iturria-Medina Y. Distinctive whole-brain cell types predict tissue damage patterns in thirteen neurodegenerative conditions. eLife 2024; 12:RP89368. [PMID: 38512130 PMCID: PMC10957173 DOI: 10.7554/elife.89368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2024] Open
Abstract
For over a century, brain research narrative has mainly centered on neuron cells. Accordingly, most neurodegenerative studies focus on neuronal dysfunction and their selective vulnerability, while we lack comprehensive analyses of other major cell types' contribution. By unifying spatial gene expression, structural MRI, and cell deconvolution, here we describe how the human brain distribution of canonical cell types extensively predicts tissue damage in 13 neurodegenerative conditions, including early- and late-onset Alzheimer's disease, Parkinson's disease, dementia with Lewy bodies, amyotrophic lateral sclerosis, mutations in presenilin-1, and 3 clinical variants of frontotemporal lobar degeneration (behavioral variant, semantic and non-fluent primary progressive aphasia) along with associated three-repeat and four-repeat tauopathies and TDP43 proteinopathies types A and C. We reconstructed comprehensive whole-brain reference maps of cellular abundance for six major cell types and identified characteristic axes of spatial overlapping with atrophy. Our results support the strong mediating role of non-neuronal cells, primarily microglia and astrocytes, in spatial vulnerability to tissue loss in neurodegeneration, with distinct and shared across-disorder pathomechanisms. These observations provide critical insights into the multicellular pathophysiology underlying spatiotemporal advance in neurodegeneration. Notably, they also emphasize the need to exceed the current neuro-centric view of brain diseases, supporting the imperative for cell-specific therapeutic targets in neurodegeneration.
Collapse
Affiliation(s)
- Veronika Pak
- Department of Neurology and Neurosurgery, McGill UniversityMontrealCanada
- McConnell Brain Imaging Centre, Montreal Neurological InstituteMontrealCanada
- Ludmer Centre for Neuroinformatics & Mental HealthMontrealCanada
| | - Quadri Adewale
- Department of Neurology and Neurosurgery, McGill UniversityMontrealCanada
- McConnell Brain Imaging Centre, Montreal Neurological InstituteMontrealCanada
- Ludmer Centre for Neuroinformatics & Mental HealthMontrealCanada
| | - Danilo Bzdok
- McConnell Brain Imaging Centre, Montreal Neurological InstituteMontrealCanada
- Department of Biomedical Engineering, McGill UniversityMontrealCanada
- School of Computer Science, McGill UniversityMontrealCanada
- Mila – Quebec Artificial Intelligence InstituteMontrealCanada
| | | | | | - Yasser Iturria-Medina
- Department of Neurology and Neurosurgery, McGill UniversityMontrealCanada
- McConnell Brain Imaging Centre, Montreal Neurological InstituteMontrealCanada
- Ludmer Centre for Neuroinformatics & Mental HealthMontrealCanada
- Department of Biomedical Engineering, McGill UniversityMontrealCanada
- McGill Centre for Studies in AgingMontrealCanada
| |
Collapse
|
9
|
Jackson WS, Bauer S, Kaczmarczyk L, Magadi SS. Selective Vulnerability to Neurodegenerative Disease: Insights from Cell Type-Specific Translatome Studies. BIOLOGY 2024; 13:67. [PMID: 38392286 PMCID: PMC10886597 DOI: 10.3390/biology13020067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/12/2024] [Accepted: 01/17/2024] [Indexed: 02/24/2024]
Abstract
Neurodegenerative diseases (NDs) manifest a wide variety of clinical symptoms depending on the affected brain regions. Gaining insights into why certain regions are resistant while others are susceptible is vital for advancing therapeutic strategies. While gene expression changes offer clues about disease responses across brain regions, the mixture of cell types therein obscures experimental results. In recent years, methods that analyze the transcriptomes of individual cells (e.g., single-cell RNA sequencing or scRNAseq) have been widely used and have provided invaluable insights into specific cell types. Concurrently, transgene-based techniques that dissect cell type-specific translatomes (CSTs) in model systems, like RiboTag and bacTRAP, offer unique advantages but have received less attention. This review juxtaposes the merits and drawbacks of both methodologies, focusing on the use of CSTs in understanding conditions like amyotrophic lateral sclerosis (ALS), Huntington's disease (HD), Alzheimer's disease (AD), and specific prion diseases like fatal familial insomnia (FFI), genetic Creutzfeldt-Jakob disease (gCJD), and acquired prion disease. We conclude by discussing the emerging trends observed across multiple diseases and emerging methods.
Collapse
Affiliation(s)
- Walker S Jackson
- Wallenberg Center for Molecular Medicine, Linköping University, 581 85 Linköping, Sweden
- Department of Biomedical and Clinical Sciences, Linköping University, 581 85 Linköping, Sweden
| | - Susanne Bauer
- Wallenberg Center for Molecular Medicine, Linköping University, 581 85 Linköping, Sweden
- Department of Biomedical and Clinical Sciences, Linköping University, 581 85 Linköping, Sweden
| | - Lech Kaczmarczyk
- Wallenberg Center for Molecular Medicine, Linköping University, 581 85 Linköping, Sweden
- Department of Biomedical and Clinical Sciences, Linköping University, 581 85 Linköping, Sweden
| | - Srivathsa S Magadi
- Wallenberg Center for Molecular Medicine, Linköping University, 581 85 Linköping, Sweden
- Department of Biomedical and Clinical Sciences, Linköping University, 581 85 Linköping, Sweden
| |
Collapse
|
10
|
Jackson WS. Etiology matters: genetic and acquired prion diseases engage different mechanisms at a presymptomatic stage. Neural Regen Res 2023; 18:2707-2708. [PMID: 37449633 DOI: 10.4103/1673-5374.373684] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023] Open
Affiliation(s)
- Walker S Jackson
- Department of Biomedical and Clinical Sciences, Wallenberg Center for Molecular Medicine, Linköping University, Linköping, Sweden
| |
Collapse
|
11
|
Jung R, Lechler MC, Fernandez-Villegas A, Chung CW, Jones HC, Choi YH, Thompson MA, Rödelsperger C, Röseler W, Kaminski Schierle GS, Sommer RJ, David DC. A safety mechanism enables tissue-specific resistance to protein aggregation during aging in C. elegans. PLoS Biol 2023; 21:e3002284. [PMID: 37708127 PMCID: PMC10501630 DOI: 10.1371/journal.pbio.3002284] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 08/01/2023] [Indexed: 09/16/2023] Open
Abstract
During aging, proteostasis capacity declines and distinct proteins become unstable and can accumulate as protein aggregates inside and outside of cells. Both in disease and during aging, proteins selectively aggregate in certain tissues and not others. Yet, tissue-specific regulation of cytoplasmic protein aggregation remains poorly understood. Surprisingly, we found that the inhibition of 3 core protein quality control systems, namely chaperones, the proteasome, and macroautophagy, leads to lower levels of age-dependent protein aggregation in Caenorhabditis elegans pharyngeal muscles, but higher levels in body-wall muscles. We describe a novel safety mechanism that selectively targets newly synthesized proteins to suppress their aggregation and associated proteotoxicity. The safety mechanism relies on macroautophagy-independent lysosomal degradation and involves several previously uncharacterized components of the intracellular pathogen response (IPR). We propose that this protective mechanism engages an anti-aggregation machinery targeting aggregating proteins for lysosomal degradation.
Collapse
Affiliation(s)
- Raimund Jung
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Marie C. Lechler
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Graduate Training Centre of Neuroscience, International Max Planck Research School, Tübingen, Germany
| | - Ana Fernandez-Villegas
- Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, United Kingdom
| | - Chyi Wei Chung
- Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, United Kingdom
| | - Harry C. Jones
- The Babraham Institute, Signalling Program, Cambridge, United Kingdom
| | - Yoon Hee Choi
- The Babraham Institute, Signalling Program, Cambridge, United Kingdom
| | | | - Christian Rödelsperger
- Max Planck Institute for Developmental Biology, Department for Integrative Evolutionary Biology, Tübingen, Germany
| | - Waltraud Röseler
- Max Planck Institute for Developmental Biology, Department for Integrative Evolutionary Biology, Tübingen, Germany
| | | | - Ralf J. Sommer
- Max Planck Institute for Developmental Biology, Department for Integrative Evolutionary Biology, Tübingen, Germany
| | - Della C. David
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- The Babraham Institute, Signalling Program, Cambridge, United Kingdom
- Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen, Germany
| |
Collapse
|
12
|
Cracco L, Puoti G, Cornacchia A, Glisic K, Lee SK, Wang Z, Cohen ML, Appleby BS, Cali I. Novel histotypes of sporadic Creutzfeldt-Jakob disease linked to 129MV genotype. Acta Neuropathol Commun 2023; 11:141. [PMID: 37653534 PMCID: PMC10469800 DOI: 10.1186/s40478-023-01631-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 08/01/2023] [Indexed: 09/02/2023] Open
Abstract
The MV1 and MV2 subtypes of sporadic Creutzfeldt-Jakob disease (sCJD) are linked to the heterozygous methionine (M)/valine (V) polymorphism at codon 129 of the prion protein (PrP) gene. MV2 is phenotypically heterogeneous, whereas MV1, due to its low prevalence, is one of the least well characterized subtypes. In this study, we investigated the biochemical properties of PrPSc and phenotypic expression of cases diagnosed as sCJD MV1 and MV2. We describe four MV2 histotypes: 2C, with cortical (C) coarse pathology; 2K, with kuru (K) plaque deposits; 2C-K, with co-existing C and K histotypic features; and the novel histotype 2C-PL that mimics 2C in the cerebral cortex and cerebellum, but exhibits plaque-like (PL) PrP deposits in subcortical regions (e.g., basal nuclei, thalamus and midbrain). Histotype prevalence is highest for 2C-K (55%), intermediate for 2C (31%), and lowest for 2C-PL and 2K (7%). Nearly every MV2 case expressed both PrPSc types, with T2 being the predominant type ("MV2-1"). MV1 cases typically show a rapid disease course (≤ 4 months), and feature the 1C histotype, phenotypically identical to sCJDMM1. Co-existing PrPSc types, with T1 significantly exceeding T2 ("MV1-2"), are detected in patients diagnosed as MV1 with longer disease courses. We observed four histotypes among MV1-2 cases, including two novel histotypes: 1V, reminiscent of sCJDVV1; 1C-2C, resembling sCJDMM1-2 with predominant MM1 histotypic component; and novel histotypes 1C-2PL and 1C-2K, overall mimicking 1C in the cerebral cortex, but harboring T2 and plaque-like PrP deposits in subcortical regions (1C-2PL), and T2 and kuru plaques in the cerebellum (1C-2K). Lesion profiles of 1C, 1V, and 1C-2C are similar, but differ from 1C-2PL and 1C-2K, as the latter two groups show prominent hippocampal and nigral degeneration. We believe that the novel "C-PL" histotypes are distinct entities rather than intermediate forms between "C" and "C-K" groups, and that 1C-2PL and 1C-2K histotypes may be characterized by different T1 variants of the same size.
Collapse
Affiliation(s)
- Laura Cracco
- Department of Pathology and Laboratory Medicine, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA
| | - Gianfranco Puoti
- Division of Neurology, University of Campania "Luigi Vanvitelli", Caserta, Italy
| | - Antonio Cornacchia
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Katie Glisic
- National Prion Disease Pathology Surveillance Center, Cleveland, OH, 44106, USA
| | - Seong-Ki Lee
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Zerui Wang
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Mark L Cohen
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
- National Prion Disease Pathology Surveillance Center, Cleveland, OH, 44106, USA
| | - Brian S Appleby
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
- National Prion Disease Pathology Surveillance Center, Cleveland, OH, 44106, USA
- Department of Neurology, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
- Department of Psychiatry, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Ignazio Cali
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA.
- National Prion Disease Pathology Surveillance Center, Cleveland, OH, 44106, USA.
| |
Collapse
|
13
|
Ellis EG, Joutsa J, Morrison-Ham J, Younger EFP, Saward JB, Caeyenberghs K, Corp DT. Large-scale activation likelihood estimation meta-analysis of parkinsonian disorders. Brain Commun 2023; 5:fcad172. [PMID: 37324240 PMCID: PMC10265724 DOI: 10.1093/braincomms/fcad172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/31/2023] [Accepted: 05/29/2023] [Indexed: 06/17/2023] Open
Abstract
Parkinsonism is a feature of several neurodegenerative disorders, including Parkinson's disease, progressive supranuclear palsy, corticobasal syndrome and multiple system atrophy. Neuroimaging studies have yielded insights into parkinsonian disorders; however, due to variability in results, the brain regions consistently implicated in these disorders remain to be characterized. The aim of this meta-analysis was to identify consistent brain abnormalities in individual parkinsonian disorders (Parkinson's disease, progressive supranuclear palsy, corticobasal syndrome and multiple system atrophy) and to investigate any shared abnormalities across disorders. A total of 44 591 studies were systematically screened following searches of two databases. A series of whole-brain activation likelihood estimation meta-analyses were performed on 132 neuroimaging studies (69 Parkinson's disease; 23 progressive supranuclear palsy; 17 corticobasal syndrome; and 23 multiple system atrophy) utilizing anatomical MRI, perfusion or metabolism PET and single-photon emission computed tomography. Meta-analyses were performed in each parkinsonian disorder within each imaging modality, as well as across all included disorders. Results in progressive supranuclear palsy and multiple system atrophy aligned with current imaging markers for diagnosis, encompassing the midbrain, and brainstem and putamen, respectively. PET imaging studies of patients with Parkinson's disease most consistently reported abnormality of the middle temporal gyrus. No significant clusters were identified in corticobasal syndrome. When examining abnormalities shared across all four disorders, the caudate was consistently reported in MRI studies, whilst the thalamus, inferior frontal gyrus and middle temporal gyri were commonly implicated by PET. To our knowledge, this is the largest meta-analysis of neuroimaging studies in parkinsonian disorders and the first to characterize brain regions implicated across parkinsonian disorders.
Collapse
Affiliation(s)
- Elizabeth G Ellis
- Correspondence to: Elizabeth G. Ellis Cognitive Neuroscience Unit, School of Psychology Deakin University, 221 Burwood Highway Burwood, VIC 3125, Australia E-mail:
| | - Juho Joutsa
- Center for Brain Circuit Therapeutics, Department of Neurology, Psychiatry, and Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Turku Brain and Mind Center, Clinical Neurosciences, University of Turku, Turku 20520, Finland
- Turku PET Centre, Neurocenter, Turku University Hospital, Turku 20520, Finland
| | - Jordan Morrison-Ham
- Cognitive Neuroscience Unit, School of Psychology, Deakin University, Geelong, VIC 3220, Australia
| | - Ellen F P Younger
- Cognitive Neuroscience Unit, School of Psychology, Deakin University, Geelong, VIC 3220, Australia
| | - Jacqueline B Saward
- Cognitive Neuroscience Unit, School of Psychology, Deakin University, Geelong, VIC 3220, Australia
| | - Karen Caeyenberghs
- Cognitive Neuroscience Unit, School of Psychology, Deakin University, Geelong, VIC 3220, Australia
| | - Daniel T Corp
- Correspondence may also be addressed to: Daniel T. Corp Cognitive Neuroscience Unit, School of Psychology Deakin University, 221 Burwood Highway Burwood, VIC 3125, Australia E-mail:
| |
Collapse
|
14
|
Walters RO, Haigh CL. Organoids for modeling prion diseases. Cell Tissue Res 2023; 392:97-111. [PMID: 35088182 PMCID: PMC9329493 DOI: 10.1007/s00441-022-03589-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 01/19/2022] [Indexed: 11/25/2022]
Abstract
Human cerebral organoids are an exciting and novel model system emerging in the field of neurobiology. Cerebral organoids are spheres of self-organizing, neuronal lineage tissue that can be differentiated from human pluripotent stem cells and that present the possibility of on-demand human neuronal cultures that can be used for non-invasively investigating diseases affecting the brain. Compared with existing humanized cell models, they provide a more comprehensive replication of the human cerebral environment. The potential of the human cerebral organoid model is only just beginning to be elucidated, but initial studies have indicated that they could prove to be a valuable model for neurodegenerative diseases such as prion disease. The application of the cerebral organoid model to prion disease, what has been learned so far and the future potential of this model are discussed in this review.
Collapse
Affiliation(s)
- Ryan O Walters
- Prion Cell Biology Unit, Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 903 South 4th Street, Hamilton, MT, 59840, USA
| | - Cathryn L Haigh
- Prion Cell Biology Unit, Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 903 South 4th Street, Hamilton, MT, 59840, USA.
| |
Collapse
|
15
|
Groveman BR, Race B, Foliaki ST, Williams K, Hughson AG, Baune C, Zanusso G, Haigh CL. Sporadic Creutzfeldt-Jakob disease infected human cerebral organoids retain the original human brain subtype features following transmission to humanized transgenic mice. Acta Neuropathol Commun 2023; 11:28. [PMID: 36788566 PMCID: PMC9930245 DOI: 10.1186/s40478-023-01512-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 01/11/2023] [Indexed: 02/16/2023] Open
Abstract
Human cerebral organoids (COs) are three-dimensional self-organizing cultures of cerebral brain tissue differentiated from induced pluripotent stem cells. We have recently shown that COs are susceptible to infection with different subtypes of Creutzfeldt-Jakob disease (CJD) prions, which in humans cause different manifestations of the disease. The ability to study live human brain tissue infected with different CJD subtypes opens a wide array of possibilities from differentiating mechanisms of cell death and identifying neuronal selective vulnerabilities to testing therapeutics. However, the question remained as to whether the prions generated in the CO model truly represent those in the infecting inoculum. Mouse models expressing human prion protein are commonly used to characterize human prion disease as they reproduce many of the molecular and clinical phenotypes associated with CJD subtypes. We therefore inoculated these mice with COs that had been infected with two CJD subtypes (MV1 and MV2) to see if the original subtype characteristics (referred to as strains once transmitted into a model organism) of the infecting prions were maintained in the COs when compared with the original human brain inocula. We found that disease characteristics caused by the molecular subtype of the disease associated prion protein were similar in mice inoculated with either CO derived material or human brain material, demonstrating that the disease associated prions generated in COs shared strain characteristics with those in humans. As the first and only in vitro model of human neurodegenerative disease that can faithfully reproduce different subtypes of prion disease, these findings support the use of the CO model for investigating human prion diseases and their subtypes.
Collapse
Affiliation(s)
- Bradley R. Groveman
- grid.419681.30000 0001 2164 9667Laboratory of Persistent Viral Diseases, Division of Intramural Research, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 903 South 4th Street, Hamilton, MT 59840 USA
| | - Brent Race
- grid.419681.30000 0001 2164 9667Laboratory of Persistent Viral Diseases, Division of Intramural Research, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 903 South 4th Street, Hamilton, MT 59840 USA
| | - Simote T. Foliaki
- grid.419681.30000 0001 2164 9667Laboratory of Persistent Viral Diseases, Division of Intramural Research, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 903 South 4th Street, Hamilton, MT 59840 USA
| | - Katie Williams
- grid.419681.30000 0001 2164 9667Laboratory of Persistent Viral Diseases, Division of Intramural Research, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 903 South 4th Street, Hamilton, MT 59840 USA
| | - Andrew G. Hughson
- grid.419681.30000 0001 2164 9667Laboratory of Persistent Viral Diseases, Division of Intramural Research, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 903 South 4th Street, Hamilton, MT 59840 USA
| | - Chase Baune
- grid.419681.30000 0001 2164 9667Laboratory of Persistent Viral Diseases, Division of Intramural Research, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 903 South 4th Street, Hamilton, MT 59840 USA
| | - Gianluigi Zanusso
- grid.5611.30000 0004 1763 1124Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Cathryn L. Haigh
- grid.419681.30000 0001 2164 9667Laboratory of Persistent Viral Diseases, Division of Intramural Research, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 903 South 4th Street, Hamilton, MT 59840 USA
| |
Collapse
|
16
|
Bauer S, Dittrich L, Kaczmarczyk L, Schleif M, Benfeitas R, Jackson WS. Translatome profiling in fatal familial insomnia implicates TOR signaling in somatostatin neurons. Life Sci Alliance 2022; 5:5/11/e202201530. [PMID: 36192034 PMCID: PMC9531780 DOI: 10.26508/lsa.202201530] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 09/09/2022] [Accepted: 09/12/2022] [Indexed: 11/24/2022] Open
Abstract
Bauer and colleagues report that among the six neuron types studied, somatostatin neurons have an unexpectedly strong and similar response to two distinct genetic prion diseases before disease onset. Selective neuronal vulnerability is common in neurodegenerative diseases but poorly understood. In genetic prion diseases, including fatal familial insomnia (FFI) and Creutzfeldt–Jakob disease (CJD), different mutations in the Prnp gene manifest as clinically and neuropathologically distinct diseases. Here we report with electroencephalography studies that theta waves are mildly increased in 21 mo old knock-in mice modeling FFI and CJD and that sleep is mildy affected in FFI mice. To define affected cell types, we analyzed cell type–specific translatomes from six neuron types of 9 mo old FFI and CJD mice. Somatostatin (SST) neurons responded the strongest in both diseases, with unexpectedly high overlap in genes and pathways. Functional analyses revealed up-regulation of neurodegenerative disease pathways and ribosome and mitochondria biogenesis, and down-regulation of synaptic function and small GTPase-mediated signaling in FFI, implicating down-regulation of mTOR signaling as the root of these changes. In contrast, responses in glutamatergic cerebellar neurons were disease-specific. The high similarity in SST neurons of FFI and CJD mice suggests that a common therapy may be beneficial for multiple genetic prion diseases.
Collapse
Affiliation(s)
- Susanne Bauer
- Department of Biomedical and Clinical Sciences, Wallenberg Center for Molecular Medicine, Linköping University, Linköping, Sweden
| | - Lars Dittrich
- German Center for Neurodegenerative Diseases, Bonn, Germany
| | - Lech Kaczmarczyk
- Department of Biomedical and Clinical Sciences, Wallenberg Center for Molecular Medicine, Linköping University, Linköping, Sweden.,German Center for Neurodegenerative Diseases, Bonn, Germany
| | - Melvin Schleif
- German Center for Neurodegenerative Diseases, Bonn, Germany
| | - Rui Benfeitas
- Department of Biochemistry and Biophysics, National Bioinformatics Infrastructure Sweden (NBIS), Science for Life Laboratory, Stockholm University, Stockholm, Sweden
| | - Walker S Jackson
- Department of Biomedical and Clinical Sciences, Wallenberg Center for Molecular Medicine, Linköping University, Linköping, Sweden .,German Center for Neurodegenerative Diseases, Bonn, Germany
| |
Collapse
|
17
|
Kaczmarczyk L, Schleif M, Dittrich L, Williams RH, Koderman M, Bansal V, Rajput A, Schulte T, Jonson M, Krost C, Testaquadra FJ, Bonn S, Jackson WS. Distinct translatome changes in specific neural populations precede electroencephalographic changes in prion-infected mice. PLoS Pathog 2022; 18:e1010747. [PMID: 35960762 PMCID: PMC9401167 DOI: 10.1371/journal.ppat.1010747] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 08/24/2022] [Accepted: 07/18/2022] [Indexed: 12/04/2022] Open
Abstract
Selective vulnerability is an enigmatic feature of neurodegenerative diseases (NDs), whereby a widely expressed protein causes lesions in specific cell types and brain regions. Using the RiboTag method in mice, translational responses of five neural subtypes to acquired prion disease (PrD) were measured. Pre-onset and disease onset timepoints were chosen based on longitudinal electroencephalography (EEG) that revealed a gradual increase in theta power between 10- and 18-weeks after prion injection, resembling a clinical feature of human PrD. At disease onset, marked by significantly increased theta power and histopathological lesions, mice had pronounced translatome changes in all five cell types despite appearing normal. Remarkably, at a pre-onset stage, prior to EEG and neuropathological changes, we found that 1) translatomes of astrocytes indicated reduced synthesis of ribosomal and mitochondrial components, 2) glutamatergic neurons showed increased expression of cytoskeletal genes, and 3) GABAergic neurons revealed reduced expression of circadian rhythm genes. These data demonstrate that early translatome responses to neurodegeneration emerge prior to conventional markers of disease and are cell type-specific. Therapeutic strategies may need to target multiple pathways in specific populations of cells, early in disease. Prions are infectious agents composed of a misfolded protein. When isolated from a mammalian brain and transferred to the same host species, prions will cause the same neurodegenerative disease affecting the same brain regions and cell types. This concept of selective vulnerability is also a feature of more common types of neurodegenerative diseases, such as Alzheimer’s, Parkinson’s, and Huntington’s. To better understand the mechanisms behind selective vulnerability, we studied disease responses of five cell types with different vulnerabilities in prion-infected mice at two different disease stages. Responses were measured as changes to mRNAs undergoing translation, referred to as the translatome. Before prion-infected mice demonstrated typical disease signs, electroencephalography (a method used clinically to characterize neurodegeneration in humans) revealed brain changes resembling those in human prion diseases, and surprisingly, the translatomes of all cells were drastically changed. Furthermore, before electroencephalography changes emerged, three cell types made unique responses while the most vulnerable cell type did not. These results suggests that mechanisms causing selective vulnerability will be difficult to dissect and that therapies will likely need to be provided before clinical signs emerge and individually engage multiple cell types and their distinct molecular pathways.
Collapse
Affiliation(s)
- Lech Kaczmarczyk
- Wallenberg Center for Molecular Medicine, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- German Center for Neurodegenerative Diseases, Bonn, Germany
| | - Melvin Schleif
- German Center for Neurodegenerative Diseases, Bonn, Germany
| | - Lars Dittrich
- German Center for Neurodegenerative Diseases, Bonn, Germany
| | | | - Maruša Koderman
- Wallenberg Center for Molecular Medicine, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Vikas Bansal
- Institute of Medical Systems Biology, Center for Biomedical AI (bAIome), Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg-Eppendorf, Germany
- German Center for Neurodegenerative Diseases, Tübingen, Germany
| | - Ashish Rajput
- Institute of Medical Systems Biology, Center for Biomedical AI (bAIome), Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg-Eppendorf, Germany
- Maximon AG, Zug, Switzerland
| | | | - Maria Jonson
- Wallenberg Center for Molecular Medicine, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Clemens Krost
- German Center for Neurodegenerative Diseases, Bonn, Germany
| | | | - Stefan Bonn
- Institute of Medical Systems Biology, Center for Biomedical AI (bAIome), Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg-Eppendorf, Germany
| | - Walker S. Jackson
- Wallenberg Center for Molecular Medicine, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- German Center for Neurodegenerative Diseases, Bonn, Germany
- * E-mail:
| |
Collapse
|
18
|
Pandya S, Maia PD, Freeze B, Menke RAL, Talbot K, Turner MR, Raj A. Modeling seeding and neuroanatomic spread of pathology in amyotrophic lateral sclerosis. Neuroimage 2022; 251:118968. [PMID: 35143975 PMCID: PMC10729776 DOI: 10.1016/j.neuroimage.2022.118968] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 01/26/2022] [Accepted: 02/02/2022] [Indexed: 12/12/2022] Open
Abstract
The neurodegenerative disorder amyotrophic lateral sclerosis (ALS) is characterized by the progressive loss of upper and lower motor neurons, with pathological involvement of cerebral motor and extra-motor areas in a clinicopathological spectrum with frontotemporal dementia (FTD). A key unresolved issue is how the non-random distribution of pathology in ALS reflects differential network vulnerability, including molecular factors such as regional gene expression, or preferential spread of pathology via anatomical connections. A system of histopathological staging of ALS based on the regional burden of TDP-43 pathology observed in postmortem brains has been supported to some extent by analysis of distribution of in vivo structural MRI changes. In this paper, computational modeling using a Network Diffusion Model (NDM) was used to investigate whether a process of focal pathological 'seeding' followed by structural network-based spread recapitulated postmortem histopathological staging and, secondly, whether this had any correlation to the pattern of expression of a panel of genes implicated in ALS across the healthy brain. Regionally parcellated T1-weighted MRI data from ALS patients (baseline n=79) was studied in relation to a healthy control structural connectome and a database of associated regional cerebral gene expression. The NDM provided strong support for a structural network-based basis for regional pathological spread in ALS, but no simple relationship to the spatial distribution of ALS-related genes in the healthy brain. Interestingly, OPTN gene was identified as a significant but a weaker non-NDM contributor within the network-gene interaction model (LASSO). Intriguingly, the critical seed regions for spread within the model were not within the primary motor cortex but basal ganglia, thalamus and insula, where NDM recapitulated aspects of the postmortem histopathological staging system. Within the ALS-FTD clinicopathological spectrum, non-primary motor structures may be among the earliest sites of cerebral pathology.
Collapse
Affiliation(s)
- Sneha Pandya
- Department of Radiology, Weill Cornell Medicine, 1300 York Avenue, New York, NY, United States.
| | - Pedro D Maia
- Department of Mathematics, University of Texas at Arlington, TX, United States
| | - Benjamin Freeze
- Scripps Health/MD Anderson Cancer Center, Department of Radiology, CA, United States
| | - Ricarda A L Menke
- Wellcome Centre for Integrative Neuroimaging, University of Oxford, West Wing Level 6, Oxford OX2 7PZ, United Kingdom
| | - Kevin Talbot
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Martin R Turner
- Wellcome Centre for Integrative Neuroimaging, University of Oxford, West Wing Level 6, Oxford OX2 7PZ, United Kingdom; Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom.
| | - Ashish Raj
- Department of Radiology, Weill Cornell Medicine, 1300 York Avenue, New York, NY, United States; Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA 94121, United States.
| |
Collapse
|
19
|
Wang X, Liu EJ, Liu Q, Li SH, Li T, Zhou QZ, Liu YC, Zhang H, Wang JZ. Tau Acetylation in Entorhinal Cortex Induces its Chronic Hippocampal Propagation and Cognitive Deficits in Mice. J Alzheimers Dis 2021; 77:241-255. [PMID: 32804150 DOI: 10.3233/jad-200529] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Increased tau acetylation at K174, K274, K280, and K281 has been observed in the brains of Alzheimer's disease (AD) patients or in transgenic mice, but the role of acetylation in tau propagation is elusive. OBJECTIVE To study the effect of tau acetylation in entorhinal cortex on tau transmission and learning and memory. METHODS Stereotactic brain injection, behavioral test, electrophysiological recording, immunohistochemistry, and immunofluorescence were used. RESULTS We constructed the hyperacetylation mimics of tau (AAV-Tau-4Q), the non-acetylation tau mutant (AAV-Tau-4R), and the wild-type tau (AAV-Tau-WT). By overexpressing these different tau proteins in the entorhinal cortex (EC) of 2-month-old mice, we found that overexpressing Tau-4Q in EC for 3 or 6 months (to 5 or 8 months of age) neither induces tau propagation to dentate gyrus (DG) nor glial activation in DG, nor spatial memory deficit. However, overexpressing Tau-WT and Tau-4Q in EC for 13.5 months (15.5 months of age) at 2 months promoted tau propagation respectively to granulosa and hilus of DG with glial activation, synaptic dysfunction, and memory deficit, while overexpressing Tau-4R abolished tau propagation with improved cellular pathologies and cognitive functions. Furthermore, overexpressing Tau-4Q in unilateral DG of 2-month-old mice for 8 weeks also promoted its contralateral transmission with glial activation, and mice with tau (Tau-WT, Tau-4Q, and Tau-4R) overexpression in DG showed cognitive deficits compared with the empty vector controls. CONCLUSION Tau acetylation induces a time-dependent propagation from EC to DG, and only hippocampus but not EC tau accumulation induces cognitive deficits.
Collapse
Affiliation(s)
- Xin Wang
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - En-Jie Liu
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qian Liu
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shi-Hong Li
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ting Li
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiu-Zhi Zhou
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan-Chao Liu
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huaqiu Zhang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian-Zhi Wang
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
20
|
Lee MTW, Mahy W, Rackham MD. The medicinal chemistry of mitochondrial dysfunction: a critical overview of efforts to modulate mitochondrial health. RSC Med Chem 2021; 12:1281-1311. [PMID: 34458736 PMCID: PMC8372206 DOI: 10.1039/d1md00113b] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 05/17/2021] [Indexed: 12/16/2022] Open
Abstract
Mitochondria are subcellular organelles that perform a variety of critical biological functions, including ATP production and acting as hubs of immune and apoptotic signalling. Mitochondrial dysfunction has been extensively linked to the pathology of multiple neurodegenerative disorders, resulting in significant investment from the drug discovery community. Despite extensive efforts, there remains no disease modifying therapies for neurodegenerative disorders. This manuscript aims to review the compounds historically used to modulate the mitochondrial network through the lens of modern medicinal chemistry, and to offer a perspective on the evidence that relevant exposure was achieved in a representative model and that exposure was likely to result in target binding and engagement of pharmacology. We hope this manuscript will aid the community in identifying those targets and mechanisms which have been convincingly (in)validated with high quality chemical matter, and those for which an opportunity exists to explore in greater depth.
Collapse
Affiliation(s)
| | - William Mahy
- MSD The Francis Crick Institute 1 Midland Road London NW1 1AT UK
| | | |
Collapse
|
21
|
Raj A. Graph Models of Pathology Spread in Alzheimer's Disease: An Alternative to Conventional Graph Theoretic Analysis. Brain Connect 2021; 11:799-814. [PMID: 33858198 DOI: 10.1089/brain.2020.0905] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Background: Graph theory and connectomics are new techniques for uncovering disease-induced changes in the brain's structural network. Most prior studied have focused on network statistics as biomarkers of disease. However, an emerging body of work involves exploring how the network serves as a conduit for the propagation of disease factors in the brain and has successfully mapped the functional and pathological consequences of disease propagation. In Alzheimer's disease (AD), progressive deposition of misfolded proteins amyloid and tau is well-known to follow fiber projections, under a "prion-like" trans-neuronal transmission mechanism, through which misfolded proteins cascade along neuronal pathways, giving rise to network spread. Methods: In this review, we survey the state of the art in mathematical modeling of connectome-mediated pathology spread in AD. Then we address several open questions that are amenable to mathematically precise parsimonious modeling of pathophysiological processes, extrapolated to the whole brain. We specifically identify current formal models of how misfolded proteins are produced, aggregate, and disseminate in brain circuits, and attempt to understand how this process leads to stereotyped progression in Alzheimer's and other related diseases. Conclusion: This review serves to unify current efforts in modeling of AD progression that together have the potential to explain observed phenomena and serve as a test-bed for future hypothesis generation and testing in silico. Impact statement Graph theory is a powerful new approach that is transforming the study of brain processes. There do not exist many focused reviews of the subfield of graph modeling of how Alzheimer's and other dementias propagate within the brain network, and how these processes can be mapped mathematically. By providing timely and topical review of this subfield, we fill a critical gap in the community and present a unified view that can serve as an in silico test-bed for future hypothesis generation and testing. We also point to several open and unaddressed questions and controversies that future practitioners can tackle.
Collapse
Affiliation(s)
- Ashish Raj
- Department of Radiology and Biomedical Imaging, University of California at San Francisco, San Francisco, California, USA
| |
Collapse
|
22
|
Fan C, Chen ZQ, Li C, Wang YL, Yu Q, Zhu MQ. Hydrophilic AIE-Active Tetraarylethenes for Fluorescence Sensing and Super-Resolution Imaging of Amyloid Fibrils from Hen Egg White Lysozyme. ACS APPLIED MATERIALS & INTERFACES 2021; 13:19625-19632. [PMID: 33886270 DOI: 10.1021/acsami.1c01819] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Hen egg white lysozyme (HEWL) is frequently applied as a model protein for research on protein folding, unfolding, and fibrillization identified by featured fluorescent probes. Here, a series of hydrophilic, pH-sensitive tetraarylethene (TAE)-type AIEgens are synthesized via a geminal cross-coupling (GCC) reaction and evaluated for their capabilities of fluorescence sensing and super-resolution localization imaging of HEWL fibrils. With superior optical and sensing properties, the selected TAE-type AIEgen probe is weakly emissive in aqueous media, without dependence on the pH value and buffer concentration, but exhibits "turn-on" fluorescence upon interaction with HEWL amyloid fibrils in a spontaneous and reversible way that just meets the requirement of fluorescence random switching for super-resolution imaging. The selected probe has the strongest fluorescence response to HEWL amyloid fibrils exhibiting a limit of detection of 0.59 nmol/L and enables super-resolution fluorescence imaging of amyloid aggregates with a high resolution of 40 nm.
Collapse
Affiliation(s)
- Cheng Fan
- Wuhan National Laboratory for Optoelectronics, School of Optical and Electronic Information, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Ze-Qiang Chen
- Wuhan National Laboratory for Optoelectronics, School of Optical and Electronic Information, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Chong Li
- Wuhan National Laboratory for Optoelectronics, School of Optical and Electronic Information, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
- Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, South China University of Technology, Guangzhou 510640, China
| | - Ya-Long Wang
- School of Biomedical Engineering, Hainan University, Haikou, Hainan 570228, China
| | - Qi Yu
- Wuhan National Laboratory for Optoelectronics, School of Optical and Electronic Information, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Ming-Qiang Zhu
- Wuhan National Laboratory for Optoelectronics, School of Optical and Electronic Information, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
- School of Biomedical Engineering, Hainan University, Haikou, Hainan 570228, China
| |
Collapse
|
23
|
Valderhaug VD, Heiney K, Ramstad OH, Bråthen G, Kuan WL, Nichele S, Sandvig A, Sandvig I. Early functional changes associated with alpha-synuclein proteinopathy in engineered human neural networks. Am J Physiol Cell Physiol 2021; 320:C1141-C1152. [PMID: 33950697 DOI: 10.1152/ajpcell.00413.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
A patterned spread of proteinopathy represents a common characteristic of many neurodegenerative diseases. In Parkinson's disease (PD), misfolded forms of α-synuclein proteins accumulate in hallmark pathological inclusions termed Lewy bodies and Lewy neurites. Such protein aggregates seem to affect selectively vulnerable neuronal populations in the substantia nigra and to propagate within interconnected neuronal networks. Research findings suggest that these proteinopathic inclusions are present at very early time points in disease development, even before clear behavioral symptoms of dysfunction arise. In this study, we investigate the early pathophysiology developing after induced formation of such PD-related α-synuclein inclusions in a physiologically relevant in vitro setup using engineered human neural networks. We monitor the neural network activity using multielectrode arrays (MEAs) for a period of 3 wk following proteinopathy induction to identify associated changes in network function, with a special emphasis on the measure of network criticality. Self-organized criticality represents the critical point between resilience against perturbation and adaptational flexibility, which appears to be a functional trait in self-organizing neural networks, both in vitro and in vivo. We show that although developing pathology at early onset is not clearly manifest in standard measurements of network function, it may be discerned by investigating differences in network criticality states.
Collapse
Affiliation(s)
- Vibeke D Valderhaug
- Department of Neuromedicine and Movement Science, Faculty of Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Kristine Heiney
- Department of Computer Science, Faculty of Technology, Art and Design, Oslo Metropolitan University (OsloMet), Oslo, Norway
| | - Ola Huse Ramstad
- Department of Neuromedicine and Movement Science, Faculty of Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Geir Bråthen
- Department of Neuromedicine and Movement Science, Faculty of Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Wei-Li Kuan
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Stefano Nichele
- Department of Computer Science, Faculty of Technology, Art and Design, Oslo Metropolitan University (OsloMet), Oslo, Norway
| | - Axel Sandvig
- Department of Neuromedicine and Movement Science, Faculty of Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.,Department of Neurology and Clinical Neurophysiology, St Olav's Hospital, Trondheim, Norway.,Department of Clinical Neurosciences, Umeå University Hospital, Umeå, Sweden.,Department of Rehabilitation Medicine, Umeå University Hospital, Umeå, Sweden.,Department of Community Medicine and Rehabilitation, Umeå University, Umeå, Sweden.,Clinical Sciences, Umeå University, Umeå, Sweden
| | - Ioanna Sandvig
- Department of Neuromedicine and Movement Science, Faculty of Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| |
Collapse
|
24
|
Barazesh M, Mohammadi S, Bahrami Y, Mokarram P, Morowvat MH, Saidijam M, Karimipoor M, Kavousipour S, Vosoughi AR, Khanaki K. CRISPR/Cas9 Technology as a Modern Genetic Manipulation Tool for Recapitulating of Neurodegenerative Disorders in Large Animal Models. Curr Gene Ther 2021; 21:130-148. [PMID: 33319680 DOI: 10.2174/1566523220666201214115024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 11/12/2020] [Accepted: 11/23/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Neurodegenerative diseases are often the consequence of alterations in structures and functions of the Central Nervous System (CNS) in patients. Despite obtaining massive genomic information concerning the molecular basis of these diseases and since the neurological disorders are multifactorial, causal connections between pathological pathways at the molecular level and CNS disorders development have remained obscure and need to be elucidated to a great extent. OBJECTIVE Animal models serve as accessible and valuable tools for understanding and discovering the roles of causative factors in the development of neurodegenerative disorders and finding appropriate treatments. Contrary to rodents and other small animals, large animals, especially non-human primates (NHPs), are remarkably similar to humans; hence, they establish suitable models for recapitulating the main human's neuropathological manifestations that may not be seen in rodent models. In addition, they serve as useful models to discover effective therapeutic targets for neurodegenerative disorders due to their similarity to humans in terms of physiology, evolutionary distance, anatomy, and behavior. METHODS In this review, we recommend different strategies based on the CRISPR-Cas9 system for generating animal models of human neurodegenerative disorders and explaining in vivo CRISPR-Cas9 delivery procedures that are applied to disease models for therapeutic purposes. RESULTS With the emergence of CRISPR/Cas9 as a modern specific gene-editing technology in the field of genetic engineering, genetic modification procedures such as gene knock-in and knock-out have become increasingly easier compared to traditional gene targeting techniques. Unlike the old techniques, this versatile technology can efficiently generate transgenic large animal models without the need to complicate lab instruments. Hence, these animals can accurately replicate the signs of neurodegenerative disorders. CONCLUSION Preclinical applications of CRISPR/Cas9 gene-editing technology supply a unique opportunity to establish animal models of neurodegenerative disorders with high accuracy and facilitate perspectives for breakthroughs in the research on the nervous system disease therapy and drug discovery. Furthermore, the useful outcomes of CRISPR applications in various clinical phases are hopeful for their translation to the clinic in a short time.
Collapse
Affiliation(s)
- Mahdi Barazesh
- School of Paramedical, Gerash University of Medical Sciences, Gerash, Iran
| | - Shiva Mohammadi
- Department of Medical Biotechnology, School of Medicine, Lorestan University of Medical Sciences, Khoram Abad, Iran
| | - Yadollah Bahrami
- Molecular Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Pooneh Mokarram
- Autophagy Research center, Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Massoud Saidijam
- Department of Molecular Medicine and Genetics, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Morteza Karimipoor
- Molecular Medicine Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Soudabeh Kavousipour
- Molecular Medicine Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Amir Reza Vosoughi
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada
| | - Korosh Khanaki
- Medical Biotechnology Research Center, Paramedicine Faculty, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
25
|
Raj A, Powell F. Network model of pathology spread recapitulates neurodegeneration and selective vulnerability in Huntington's Disease. Neuroimage 2021; 235:118008. [PMID: 33789134 DOI: 10.1016/j.neuroimage.2021.118008] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 03/16/2021] [Accepted: 03/23/2021] [Indexed: 12/12/2022] Open
Abstract
Huntington's Disease (HD), an autosomal dominant genetic disorder caused by a mutation in the Huntingtin gene (HTT), displays a stereotyped topography in the human brain and a stereotyped progression, initially appearing in the striatum. Like other degenerative diseases, spatial topography of HD is divorced from where implicated genes are expressed, a dissociation whose mechanistic underpinning is not currently understood. Cell autonomous molecular factors characterized by gene expression signatures, including proteolytic and post translational modifications, play a role in vulnerability to disease. Non-autonomous mechanisms, likely involving the brain's anatomic or functional connectivity patterns, might also be responsible for selective vulnerability in HD. Leveraging a large dataset of 635 subjects from a multinational study, this paper tests various cell-autonomous and non-autonomous models that can explain HD topography. We test whether the expression patterns of implicated genes is sufficient to explain regional HD atrophy, or whether the network transmission of protein products is required to explain them. We find that network models are capable of predicting, to a high degree, observed atrophy in human subjects. Lastly, we propose a model of anterograde network transmission, and show that it is the most parsimonious yet most likely to explain observed atrophy patterns in HD. Collectively, these data indicate that pathology spread in HD may be mediated by the brain's intrinsic structural network organization. This is the first study to systematically and quantitatively test multiple hypotheses of pathology spread in living human subjects with HD.
Collapse
Affiliation(s)
- Ashish Raj
- Department of Radiology and Biomedical Imaging, University of California at San Francisco, USA; UCSF-UC Berkeley Graduate Program in BioEngineering, University of California at San Francisco, USA; Department of Radiology, Weill Cornell Medical College of Cornell University, 407 E. 61 Street, RR106, New York, NY 10065, USA.
| | - Fon Powell
- Department of Radiology, Weill Cornell Medical College of Cornell University, 407 E. 61 Street, RR106, New York, NY 10065, USA
| |
Collapse
|
26
|
Slc1a3-2A-CreERT2 mice reveal unique features of Bergmann glia and augment a growing collection of Cre drivers and effectors in the 129S4 genetic background. Sci Rep 2021; 11:5412. [PMID: 33686166 PMCID: PMC7940647 DOI: 10.1038/s41598-021-84887-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 02/22/2021] [Indexed: 12/26/2022] Open
Abstract
Genetic variation is a primary determinant of phenotypic diversity. In laboratory mice, genetic variation can be a serious experimental confounder, and thus minimized through inbreeding. However, generalizations of results obtained with inbred strains must be made with caution, especially when working with complex phenotypes and disease models. Here we compared behavioral characteristics of C57Bl/6—the strain most widely used in biomedical research—with those of 129S4. In contrast to 129S4, C57Bl/6 demonstrated high within-strain and intra-litter behavioral hyperactivity. Although high consistency would be advantageous, the majority of disease models and transgenic tools are in C57Bl/6. We recently established six Cre driver lines and two Cre effector lines in 129S4. To augment this collection, we genetically engineered a Cre line to study astrocytes in 129S4. It was validated with two Cre effector lines: calcium indicator gCaMP5g-tdTomato and RiboTag—a tool widely used to study cell type-specific translatomes. These reporters are in different genomic loci, and in both the Cre was functional and astrocyte-specific. We found that calcium signals lasted longer and had a higher amplitude in cortical compared to hippocampal astrocytes, genes linked to a single neurodegenerative disease have highly divergent expression patterns, and that ribosome proteins are non-uniformly expressed across brain regions and cell types.
Collapse
|
27
|
Konstantoulea K, Louros N, Rousseau F, Schymkowitz J. Heterotypic interactions in amyloid function and disease. FEBS J 2021; 289:2025-2046. [PMID: 33460517 DOI: 10.1111/febs.15719] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 01/07/2021] [Accepted: 01/15/2021] [Indexed: 11/27/2022]
Abstract
Amyloid aggregation results from the self-assembly of identical aggregation-prone sequences into cross-beta-sheet structures. The process is best known for its association with a wide range of human pathologies but also as a functional mechanism in all kingdoms of life. Less well elucidated is the role of heterotypic interactions between amyloids and other proteins and macromolecules and how this contributes to disease. We here review current data with a focus on neurodegenerative amyloid-associated diseases. Evidence indicates that heterotypic interactions occur in a wide range of amyloid processes and that these interactions modify fundamental aspects of amyloid aggregation including seeding, aggregation rates and toxicity. More work is required to understand the mechanistic origin of these interactions, but current understanding suggests that both supersaturation and sequence-specific binding can contribute to heterotypic amyloid interactions. Further unravelling these mechanisms may help to answer outstanding questions in the field including the selective vulnerability of cells types and tissues and the stereotypical spreading patterns of amyloids in disease.
Collapse
Affiliation(s)
- Katerina Konstantoulea
- VIB Center for Brain and Disease Research, Leuven, Belgium.,Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Nikolaos Louros
- VIB Center for Brain and Disease Research, Leuven, Belgium.,Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Frederic Rousseau
- VIB Center for Brain and Disease Research, Leuven, Belgium.,Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Joost Schymkowitz
- VIB Center for Brain and Disease Research, Leuven, Belgium.,Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
28
|
Regional Differences in Neuroinflammation-Associated Gene Expression in the Brain of Sporadic Creutzfeldt-Jakob Disease Patients. Int J Mol Sci 2020; 22:ijms22010140. [PMID: 33375642 PMCID: PMC7795938 DOI: 10.3390/ijms22010140] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 01/15/2023] Open
Abstract
Neuroinflammation is an essential part of neurodegeneration. Yet, the current understanding of neuroinflammation-associated molecular events in distinct brain regions of prion disease patients is insufficient to lay the ground for effective treatment strategies targeting this complex neuropathological process. To address this problem, we analyzed the expression of 800 neuroinflammation-associated genes to create a profile of biological processes taking place in the frontal cortex and cerebellum of patients who suffered from sporadic Creutzfeldt-Jakob disease. The analysis was performed using NanoString nCounter technology with human neuroinflammation panel+. The observed gene expression patterns were regionally and sub-regionally distinct, suggesting a variable neuroinflammatory response. Interestingly, the observed differences could not be explained by the molecular subtypes of sporadic Creutzfeldt-Jakob disease. Furthermore, analyses of canonical pathways and upstream regulators based on differentially expressed genes indicated an overlap between biological processes taking place in different brain regions. This suggests that even smaller-scale spatial data reflecting subtle changes in brain cells' functional heterogeneity and their immediate pathologic microenvironments are needed to explain the observed differential gene expression in a greater detail.
Collapse
|
29
|
Tittelmeier J, Nachman E, Nussbaum-Krammer C. Molecular Chaperones: A Double-Edged Sword in Neurodegenerative Diseases. Front Aging Neurosci 2020; 12:581374. [PMID: 33132902 PMCID: PMC7572858 DOI: 10.3389/fnagi.2020.581374] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 09/09/2020] [Indexed: 12/16/2022] Open
Abstract
Aberrant accumulation of misfolded proteins into amyloid deposits is a hallmark in many age-related neurodegenerative diseases, including Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington’s disease (HD), and amyotrophic lateral sclerosis (ALS). Pathological inclusions and the associated toxicity appear to spread through the nervous system in a characteristic pattern during the disease. This has been attributed to a prion-like behavior of amyloid-type aggregates, which involves self-replication of the pathological conformation, intercellular transfer, and the subsequent seeding of native forms of the same protein in the neighboring cell. Molecular chaperones play a major role in maintaining cellular proteostasis by assisting the (re)-folding of cellular proteins to ensure their function or by promoting the degradation of terminally misfolded proteins to prevent damage. With increasing age, however, the capacity of this proteostasis network tends to decrease, which enables the manifestation of neurodegenerative diseases. Recently, there has been a plethora of studies investigating how and when chaperones interact with disease-related proteins, which have advanced our understanding of the role of chaperones in protein misfolding diseases. This review article focuses on the steps of prion-like propagation from initial misfolding and self-templated replication to intercellular spreading and discusses the influence that chaperones have on these various steps, highlighting both the positive and adverse consequences chaperone action can have. Understanding how chaperones alleviate and aggravate disease progression is vital for the development of therapeutic strategies to combat these debilitating diseases.
Collapse
Affiliation(s)
- Jessica Tittelmeier
- German Cancer Research Center (DKFZ), Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Eliana Nachman
- German Cancer Research Center (DKFZ), Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Carmen Nussbaum-Krammer
- German Cancer Research Center (DKFZ), Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany
| |
Collapse
|
30
|
Catania M, Di Fede G. One or more β-amyloid(s)? New insights into the prion-like nature of Alzheimer's disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 175:213-237. [PMID: 32958234 DOI: 10.1016/bs.pmbts.2020.07.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Misfolding and aggregation of proteins play a central role in the pathogenesis of several neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's and Lewy Body diseases, Frontotemporal Lobar Degeneration and prion diseases. Increasing evidence supports the view that Aβ and tau, which are the two main molecular players in AD, share with the prion protein several "prion-like" features that can be relevant for disease pathogenesis. These features essentially include structural/conformational/biochemical variations, resistance to degradation by endogenous proteases, seeding ability, attitude to form neurotoxic assemblies, spreading and propagation of toxic aggregates, transmissibility of tau- and Aβ-related pathology to animal models. Following this view, part of the recent scientific literature has generated a new reading frame for AD pathophysiology, based on the application of the prion paradigm to the amyloid cascade hypothesis in an attempt to definitely explain the key events causing the disease and inducing its occurrence under different clinical phenotypes.
Collapse
Affiliation(s)
- Marcella Catania
- Neurology 5 / Neuropathology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Giuseppe Di Fede
- Neurology 5 / Neuropathology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy.
| |
Collapse
|
31
|
Raman S, Brookhouser N, Brafman DA. Using human induced pluripotent stem cells (hiPSCs) to investigate the mechanisms by which Apolipoprotein E (APOE) contributes to Alzheimer's disease (AD) risk. Neurobiol Dis 2020; 138:104788. [PMID: 32032733 PMCID: PMC7098264 DOI: 10.1016/j.nbd.2020.104788] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 01/14/2020] [Accepted: 01/31/2020] [Indexed: 01/02/2023] Open
Abstract
Although the biochemical and pathological hallmarks of Alzheimer's disease (AD), such as axonal transport defects, synaptic loss, and selective neuronal death, are well characterized, the underlying mechanisms that cause AD are largely unknown, thereby making it difficult to design effective therapeutic interventions. Genome-wide association studies (GWAS) studies have identified several factors associated with increased AD risk. Of these genetic factors, polymorphisms in the Apolipoprotein E (APOE) gene are the strongest and most prevalent. While it has been established that the ApoE protein modulates the formation of amyloid plaques and neurofibrillary tangles, the precise molecular mechanisms by which various ApoE isoforms enhance or mitigate AD onset and progression in aging adults are yet to be elucidated. Advances in cellular reprogramming to generate disease-in-a-dish models now provide a simplified and accessible system that complements animal and primary cell models to study ApoE in the context of AD. In this review, we will describe the use and manipulation of human induced pluripotent stem cells (hiPSCs) in dissecting the interaction between ApoE and AD. First, we will provide an overview of the proposed roles that ApoE plays in modulating pathophysiology of AD. Next, we will summarize the recent studies that have employed hiPSCs to model familial and sporadic AD. Lastly, we will speculate on how current advances in genome editing technologies and organoid culture systems can be used to improve hiPSC-based tools to investigate ApoE-dependent modulation of AD onset and progression.
Collapse
Affiliation(s)
- Sreedevi Raman
- School of Biological and Health Systems Engineering, Arizona State University, United States of America
| | - Nicholas Brookhouser
- School of Biological and Health Systems Engineering, Arizona State University, United States of America; Graduate Program in Clinical Translational Sciences, University of Arizona College of Medicine-Phoenix, United States of America
| | - David A Brafman
- School of Biological and Health Systems Engineering, Arizona State University, United States of America.
| |
Collapse
|
32
|
Pestana F, Edwards-Faret G, Belgard TG, Martirosyan A, Holt MG. No Longer Underappreciated: The Emerging Concept of Astrocyte Heterogeneity in Neuroscience. Brain Sci 2020; 10:brainsci10030168. [PMID: 32183137 PMCID: PMC7139801 DOI: 10.3390/brainsci10030168] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 03/02/2020] [Accepted: 03/05/2020] [Indexed: 12/18/2022] Open
Abstract
Astrocytes are ubiquitous in the central nervous system (CNS). These cells possess thousands of individual processes, which extend out into the neuropil, interacting with neurons, other glia and blood vessels. Paralleling the wide diversity of their interactions, astrocytes have been reported to play key roles in supporting CNS structure, metabolism, blood-brain-barrier formation and control of vascular blood flow, axon guidance, synapse formation and modulation of synaptic transmission. Traditionally, astrocytes have been studied as a homogenous group of cells. However, recent studies have uncovered a surprising degree of heterogeneity in their development and function, in both the healthy and diseased brain. A better understanding of astrocyte heterogeneity is urgently needed to understand normal brain function, as well as the role of astrocytes in response to injury and disease.
Collapse
Affiliation(s)
- Francisco Pestana
- Laboratory of Glia Biology, VIB-KU Leuven Center for Brain and Disease Research, 3000 Leuven, Belgium;
| | - Gabriela Edwards-Faret
- Laboratory of Neuronal Wiring, VIB-KU Leuven Center for Brain and Disease Research, 3000 Leuven, Belgium;
- Life & Medical Sciences (LIMES) Institute, Faculty of Mathematics and Natural Sciences, University of Bonn, 53115 Bonn, Germany
| | | | - Araks Martirosyan
- Laboratory of Glia Biology, VIB-KU Leuven Center for Brain and Disease Research, 3000 Leuven, Belgium;
- Correspondence: (A.M.); (M.G.H.); Tel.: +32-16-37-31-27 (M.G.H.)
| | - Matthew G. Holt
- Laboratory of Glia Biology, VIB-KU Leuven Center for Brain and Disease Research, 3000 Leuven, Belgium;
- Leuven Brain Institute, KU Leuven, 3000 Leuven, Belgium
- Correspondence: (A.M.); (M.G.H.); Tel.: +32-16-37-31-27 (M.G.H.)
| |
Collapse
|
33
|
Rodriguez-Porcel F, Ciarlariello VB, Dwivedi AK, Lovera L, Da Prat G, Lopez-Castellanos R, Suri R, Laub H, Walker RH, Barsottini O, Pedroso JL, Espay AJ. Movement Disorders in Prionopathies: A Systematic Review. Tremor Other Hyperkinet Mov (N Y) 2019; 9:tre-09-712. [PMID: 31871824 PMCID: PMC6925394 DOI: 10.7916/tohm.v0.712] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 11/15/2019] [Indexed: 11/16/2022] Open
Abstract
Background Movement disorders are frequent features of prionopathies. However, their prevalence and onset remain poorly described. Methods We performed a systematic review of case reports and case series of pathologically- and genetically confirmed prionopathies. Timing of symptom and movement disorder onset were documented. Continuous variables were compared between two groups using the Wilcoxon rank sum test and between multiple groups using Kruskal-Wallis test. Categorical variables were compared using Fisher's exact test. Results A total of 324 cases were included in this analysis. Movement disorders were a common feature at the onset of symptoms in most prionopathies. Gait ataxia was present in more than half of cases in all types of prionopathies. The prevalence of limb ataxia (20%) and myoclonus (24%) was lower in Gerstmann-Sträussler-Scheinker disease compared to other prionopathies (p ≤ 0.004). Myoclonus was common but often a later feature in sporadic Creutzfeldt-Jakob disease (2 months before death). Chorea was uncommon but disproportionately prevalent in variant Creutzfeldt-Jakob disease (30% of cases; p < 0.001). In genetic Creutzfeldt-Jakob disease, E200K PRNP carriers exhibited gait and limb ataxia more often when compared to other mutation carriers. Discussion Movement disorders are differentially present in the course of the various prionopathies. The movement phenomenology and appearance are associated with the type of prion disease and the PRNP genotype and likely reflect the underlying pattern of neurodegeneration. Reliance on myoclonus as a diagnostic feature of sporadic Creutzfeldt-Jakob disease may delay its recognition given its relatively late appearance in the disease course.
Collapse
Affiliation(s)
- Federico Rodriguez-Porcel
- Department of Neurology, Medical University of South Carolina, Charleston, SC, USA,James J. and Joan A. Gardner Center for Parkinson Disease and Movement Disorders, Department of Neurology and Rehabilitation Medicine, University of Cincinnati, Cincinnati, OH, USA,To whom correspondence should be addressed. E-mail:
| | | | - Alok K. Dwivedi
- Division of Biostatistics & Epidemiology, Department of Molecular and Translational Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA
| | - Lilia Lovera
- Department of Neurology, Medical University of South Carolina, Charleston, SC, USA,James J. and Joan A. Gardner Center for Parkinson Disease and Movement Disorders, Department of Neurology and Rehabilitation Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Gustavo Da Prat
- Department of Neurology, Institute of Neuroscience of Buenos Aires (INEBA), Buenos Aires, AR
| | - Ricardo Lopez-Castellanos
- James J. and Joan A. Gardner Center for Parkinson Disease and Movement Disorders, Department of Neurology and Rehabilitation Medicine, University of Cincinnati, Cincinnati, OH, USA,Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Ritika Suri
- James J. and Joan A. Gardner Center for Parkinson Disease and Movement Disorders, Department of Neurology and Rehabilitation Medicine, University of Cincinnati, Cincinnati, OH, USA,Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | - Holly Laub
- James J. and Joan A. Gardner Center for Parkinson Disease and Movement Disorders, Department of Neurology and Rehabilitation Medicine, University of Cincinnati, Cincinnati, OH, USA,Department of Neurology, Indiana University, Indianapolis, IN, USA
| | - Ruth H. Walker
- Department of Neurology, James J. Peters Veterans Affairs Medical Center, Bronx, NY, USA,Department of Neurology, Mount Sinai School of Medicine, New York, NY, USA
| | - Orlando Barsottini
- Department of Neurology, Ataxia Unit, Universidade Federal de São Paulo, São Paulo, SP, BR
| | - José Luiz Pedroso
- Department of Neurology, Ataxia Unit, Universidade Federal de São Paulo, São Paulo, SP, BR
| | - Alberto J. Espay
- James J. and Joan A. Gardner Center for Parkinson Disease and Movement Disorders, Department of Neurology and Rehabilitation Medicine, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
34
|
Španić E, Langer Horvat L, Hof PR, Šimić G. Role of Microglial Cells in Alzheimer's Disease Tau Propagation. Front Aging Neurosci 2019; 11:271. [PMID: 31636558 PMCID: PMC6787141 DOI: 10.3389/fnagi.2019.00271] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Accepted: 09/19/2019] [Indexed: 12/30/2022] Open
Abstract
Uncontrolled immune response in the brain contributes to the progression of all neurodegenerative disease, including Alzheimer's disease (AD). Recent investigations have documented the prion-like features of tau protein and the involvement of microglial changes with tau pathology. While it is still unclear what sequence of events is causal, it is likely that tau seeding potential and microglial contribution to tau propagation act together, and are essential for the development and progression of degenerative changes. Based on available evidence, targeting tau seeds and controlling some signaling pathways in a complex inflammation process could represent a possible new therapeutic approach for treating neurodegenerative diseases. Recent findings propose novel diagnostic assays and markers that may be used together with standard methods to complete and improve the diagnosis and classification of these diseases. In conclusion, a novel perspective on microglia-tau relations reveals new issues to investigate and imposes different approaches for developing therapeutic strategies for AD.
Collapse
Affiliation(s)
- Ena Španić
- Laboratory for Developmental Neuropathology, Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Lea Langer Horvat
- Laboratory for Developmental Neuropathology, Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Patrick R. Hof
- Nash Family Department of Neuroscience, Ronald M. Loeb Center for Alzheimer’s Disease, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Goran Šimić
- Laboratory for Developmental Neuropathology, Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| |
Collapse
|
35
|
Kaczmarczyk L, Bansal V, Rajput A, Rahman RU, Krzyżak W, Degen J, Poll S, Fuhrmann M, Bonn S, Jackson WS. Tagger-A Swiss army knife for multiomics to dissect cell type-specific mechanisms of gene expression in mice. PLoS Biol 2019; 17:e3000374. [PMID: 31393866 PMCID: PMC6701817 DOI: 10.1371/journal.pbio.3000374] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 08/20/2019] [Accepted: 07/17/2019] [Indexed: 12/22/2022] Open
Abstract
A deep understanding of how regulation of the multiple levels of gene expression in mammalian tissues give rise to complex phenotypes has been impeded by cellular diversity. A handful of techniques were developed to tag-select nucleic acids of interest in specific cell types, thereby enabling their capture. We expanded this strategy by developing the Tagger knock-in mouse line bearing a quad-cistronic transgene combining enrichment tools for nuclei, nascent RNA, translating mRNA, and mature microRNA (miRNA). We demonstrate that Tagger can capture the desired nucleic acids, enabling multiple omics approaches to be applied to specific cell types in vivo using a single transgenic mouse line. This Methods and Resources paper describes Tagger, a knock-in mouse line bearing a quad-cistronic transgene that enables the capture of translating mRNAs, mature miRNAs, pulse-labeled total RNA, and the nucleus, all from specific cells of complex tissues.
Collapse
Affiliation(s)
- Lech Kaczmarczyk
- Wallenberg Center for Molecular Medicine, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
- German Center for Neurodegenerative Diseases, Bonn, Germany
| | - Vikas Bansal
- Institute for Medical Systems Biology, Center for Molecular Neuroscience, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ashish Rajput
- Institute for Medical Systems Biology, Center for Molecular Neuroscience, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Raza-ur Rahman
- Institute for Medical Systems Biology, Center for Molecular Neuroscience, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Wiesław Krzyżak
- Life & Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Joachim Degen
- Life & Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Stefanie Poll
- German Center for Neurodegenerative Diseases, Bonn, Germany
| | | | - Stefan Bonn
- Institute for Medical Systems Biology, Center for Molecular Neuroscience, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Neurodegenerative Diseases, Tübingen, Germany
- * E-mail: (SB); (WSJ)
| | - Walker Scot Jackson
- Wallenberg Center for Molecular Medicine, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
- German Center for Neurodegenerative Diseases, Bonn, Germany
- * E-mail: (SB); (WSJ)
| |
Collapse
|
36
|
Abstract
Prion diseases are progressive, incurable and fatal neurodegenerative conditions. The term 'prion' was first nominated to express the revolutionary concept that a protein could be infectious. We now know that prions consist of PrPSc, the pathological aggregated form of the cellular prion protein PrPC. Over the years, the term has been semantically broadened to describe aggregates irrespective of their infectivity, and the prion concept is now being applied, perhaps overenthusiastically, to all neurodegenerative diseases that involve protein aggregation. Indeed, recent studies suggest that prion diseases (PrDs) and protein misfolding disorders (PMDs) share some common disease mechanisms, which could have implications for potential treatments. Nevertheless, the transmissibility of bona fide prions is unique, and PrDs should be considered as distinct from other PMDs.
Collapse
Affiliation(s)
- Claudia Scheckel
- Institute of Neuropathology, University of Zurich, Zurich, Switzerland
| | - Adriano Aguzzi
- Institute of Neuropathology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
37
|
Pascoal TA, Mathotaarachchi S, Kang MS, Mohaddes S, Shin M, Park AY, Parent MJ, Benedet AL, Chamoun M, Therriault J, Hwang H, Cuello AC, Misic B, Soucy JP, Aston JAD, Gauthier S, Rosa-Neto P. Aβ-induced vulnerability propagates via the brain's default mode network. Nat Commun 2019; 10:2353. [PMID: 31164641 PMCID: PMC6547716 DOI: 10.1038/s41467-019-10217-w] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 04/16/2019] [Indexed: 12/17/2022] Open
Abstract
The link between brain amyloid-β (Aβ), metabolism, and dementia symptoms remains a pressing question in Alzheimer's disease. Here, using positron emission tomography ([18F]florbetapir tracer for Aβ and [18F]FDG tracer for glucose metabolism) with a novel analytical framework, we found that Aβ aggregation within the brain's default mode network leads to regional hypometabolism in distant but functionally connected brain regions. Moreover, we found that an interaction between this hypometabolism with overlapping Aβ aggregation is associated with subsequent cognitive decline. These results were also observed in transgenic Aβ rats that do not form neurofibrillary tangles, which support these findings as an independent mechanism of cognitive deterioration. These results suggest a model in which distant Aβ induces regional metabolic vulnerability, whereas the interaction between local Aβ with a vulnerable environment drives the clinical progression of dementia.
Collapse
Affiliation(s)
- Tharick A Pascoal
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, H4H 1R3, Montreal, Canada
- Montreal Neurological Institute, H3A 2B4, Montreal, Canada
| | - Sulantha Mathotaarachchi
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, H4H 1R3, Montreal, Canada
| | - Min Su Kang
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, H4H 1R3, Montreal, Canada
- Montreal Neurological Institute, H3A 2B4, Montreal, Canada
| | - Sara Mohaddes
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, H4H 1R3, Montreal, Canada
| | - Monica Shin
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, H4H 1R3, Montreal, Canada
| | - Ah Yeon Park
- Statistical Laboratory, University of Cambridge, CB3 0WB, Cambridge, UK
| | - Maxime J Parent
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, H4H 1R3, Montreal, Canada
- Montreal Neurological Institute, H3A 2B4, Montreal, Canada
| | - Andrea L Benedet
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, H4H 1R3, Montreal, Canada
| | - Mira Chamoun
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, H4H 1R3, Montreal, Canada
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, H4H 1R3, Montreal, Canada
| | - Heungsun Hwang
- Department of Psychology, McGill University, Montreal, Canada
| | - A Claudio Cuello
- Department of Pharmacology and Therapeutics, McGill University, H3A 2T5, Montreal, Canada
- Alzheimer's Disease Research Unit, The McGill University Research Centre for Studies in Aging, H4H 1R3, Montreal, Canada
| | | | | | - John A D Aston
- Statistical Laboratory, University of Cambridge, CB3 0WB, Cambridge, UK
| | - Serge Gauthier
- Alzheimer's Disease Research Unit, The McGill University Research Centre for Studies in Aging, H4H 1R3, Montreal, Canada
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, H4H 1R3, Montreal, Canada.
- Montreal Neurological Institute, H3A 2B4, Montreal, Canada.
- Department of Pharmacology and Therapeutics, McGill University, H3A 2T5, Montreal, Canada.
- Alzheimer's Disease Research Unit, The McGill University Research Centre for Studies in Aging, H4H 1R3, Montreal, Canada.
| |
Collapse
|
38
|
Ossenkoppele R, Iaccarino L, Schonhaut DR, Brown JA, La Joie R, O'Neil JP, Janabi M, Baker SL, Kramer JH, Gorno-Tempini ML, Miller BL, Rosen HJ, Seeley WW, Jagust WJ, Rabinovici GD. Tau covariance patterns in Alzheimer's disease patients match intrinsic connectivity networks in the healthy brain. Neuroimage Clin 2019; 23:101848. [PMID: 31077982 PMCID: PMC6510968 DOI: 10.1016/j.nicl.2019.101848] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 04/02/2019] [Accepted: 05/01/2019] [Indexed: 01/06/2023]
Abstract
According to the network model of neurodegeneration, the spread of pathogenic proteins occurs selectively along connected brain regions. We tested in vivo whether the distribution of filamentous tau (measured with [18F]flortaucipir-PET), fibrillar amyloid-β ([11C]PIB-PET) and glucose hypometabolism ([18F]FDG-PET) follows the intrinsic functional organization of the healthy brain. We included 63 patients with Alzheimer's disease (AD; 30 male, 63 ± 8 years) who underwent [18F]flortaucipir, [11C]PIB and [18F]FDG PET, and 1000 young adults (427 male, 21 ± 3 years) who underwent task-free fMRI. We selected six predefined disease epicenters as seeds for whole-brain voxelwise covariance analyses to compare correlated patterns of tracer uptake across AD patients against fMRI intrinsic connectivity patterns in young adults. We found a striking convergence between [18F]flortaucipir covariance patterns and intrinsic connectivity maps (range Spearman rho's: 0.32-0.78, p < .001), which corresponded with expected functional networks (range goodness-of-fit: 3.8-8.2). The topography of amyloid-β covariance patterns was more diffuse and less network-specific, while glucose hypometabolic patterns were more spatially restricted than tau but overlapped with functional networks. These findings suggest that the spatial patterns of tau and glucose hypometabolism observed in AD resemble the functional organization of the healthy brain, supporting the notion that tau pathology spreads through circumscribed brain networks and drives neurodegeneration.
Collapse
Affiliation(s)
- Rik Ossenkoppele
- Memory and Aging Center, University of California San Francisco, San Francisco, CA 94143, USA; Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA; Department of Neurology & Alzheimer Center, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam 1081 HZ, the Netherlands.
| | - Leonardo Iaccarino
- Memory and Aging Center, University of California San Francisco, San Francisco, CA 94143, USA
| | - Daniel R Schonhaut
- Memory and Aging Center, University of California San Francisco, San Francisco, CA 94143, USA; Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA
| | - Jesse A Brown
- Memory and Aging Center, University of California San Francisco, San Francisco, CA 94143, USA
| | - Renaud La Joie
- Memory and Aging Center, University of California San Francisco, San Francisco, CA 94143, USA; Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA
| | - James P O'Neil
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Mustafa Janabi
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Suzanne L Baker
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Joel H Kramer
- Memory and Aging Center, University of California San Francisco, San Francisco, CA 94143, USA
| | | | - Bruce L Miller
- Memory and Aging Center, University of California San Francisco, San Francisco, CA 94143, USA
| | - Howard J Rosen
- Memory and Aging Center, University of California San Francisco, San Francisco, CA 94143, USA
| | - William W Seeley
- Memory and Aging Center, University of California San Francisco, San Francisco, CA 94143, USA; Department of Pathology, University of California San Francisco, San Francisco, CA 94143, USA
| | - William J Jagust
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA; Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Gil D Rabinovici
- Memory and Aging Center, University of California San Francisco, San Francisco, CA 94143, USA; Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA
| |
Collapse
|
39
|
Davis S, Scott C, Ansorge O, Fischer R. Development of a Sensitive, Scalable Method for Spatial, Cell-Type-Resolved Proteomics of the Human Brain. J Proteome Res 2019; 18:1787-1795. [PMID: 30768908 PMCID: PMC6456870 DOI: 10.1021/acs.jproteome.8b00981] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
![]()
While nearly comprehensive proteome
coverage can be achieved from
bulk tissue or cultured cells, the data usually lacks spatial resolution.
As a result, tissue based proteomics averages protein abundance across
multiple cell types and/or localizations. With proteomics platforms
lacking sensitivity and throughput to undertake deep single-cell proteome
studies in order to resolve spatial or cell type dependent protein
expression gradients within tissue, proteome analysis has been combined
with sorting techniques to enrich for certain cell populations. However,
the spatial resolution and context is lost after cell sorting. Here,
we report an optimized method for the proteomic analysis of neurons
isolated from post-mortem human brain by laser capture microdissection
(LCM). We tested combinations of sample collection methods, lysis
buffers and digestion methods to maximize the number of identifications
and quantitative performance, identifying 1500 proteins from 60 000
μm2 of 10 μm thick cerebellar molecular layer
with excellent reproducibility. To demonstrate the ability of our
workflow to resolve cell type specific proteomes within human brain
tissue, we isolated sets of individual Betz and Purkinje cells. Both
neuronal cell types are involved in motor coordination and were found
to express highly specific proteomes to a depth of 2800 to 3600 proteins.
Collapse
Affiliation(s)
- Simon Davis
- Target Discovery Institute, Nuffield Department of Medicine , University of Oxford , Roosevelt Drive , Oxford , OX3 7FZ , U.K
| | - Connor Scott
- Academic Unit of Neuropathology, Nuffield Department of Clinical Neurosciences , University of Oxford, John Radcliffe Hospital , Oxford , OX3 9DU , U.K
| | - Olaf Ansorge
- Academic Unit of Neuropathology, Nuffield Department of Clinical Neurosciences , University of Oxford, John Radcliffe Hospital , Oxford , OX3 9DU , U.K
| | - Roman Fischer
- Target Discovery Institute, Nuffield Department of Medicine , University of Oxford , Roosevelt Drive , Oxford , OX3 7FZ , U.K
| |
Collapse
|
40
|
Montibeller L, de Belleroche J. Amyotrophic lateral sclerosis (ALS) and Alzheimer's disease (AD) are characterised by differential activation of ER stress pathways: focus on UPR target genes. Cell Stress Chaperones 2018; 23:897-912. [PMID: 29725981 PMCID: PMC6111088 DOI: 10.1007/s12192-018-0897-y] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 04/06/2018] [Accepted: 04/08/2018] [Indexed: 12/11/2022] Open
Abstract
The endoplasmic reticulum (ER) plays an important role in maintenance of proteostasis through the unfolded protein response (UPR), which is strongly activated in most neurodegenerative disorders. UPR signalling pathways mediated by IRE1α and ATF6 play a crucial role in the maintenance of ER homeostasis through the transactivation of an array of transcription factors. When activated, these transcription factors induce the expression of genes involved in protein folding and degradation with pro-survival effects. However, the specific contribution of these transcription factors to different neurodegenerative diseases remains poorly defined. Here, we characterised 44 target genes strongly influenced by XBP1 and ATF6 and quantified the expression of a subset of genes in the human post-mortem spinal cord from amyotrophic lateral sclerosis (ALS) cases and in the frontal and temporal cortex from frontotemporal lobar degeneration (FTLD) and Alzheimer's disease (AD) cases and controls. We found that IRE1α-XBP1 and ATF6 pathways were strongly activated both in ALS and AD. In ALS, XBP1 and ATF6 activation was confirmed by a substantial increase in the expression of both known and novel target genes involved particularly in co-chaperone activity and ER-associated degradation (ERAD) such as DNAJB9, SEL1L and OS9. In AD cases, a distinct pattern emerged, where targets involved in protein folding were more prominent, such as CANX, PDIA3 and PDIA6. These results reveal that both overlapping and disease-specific patterns of IRE1α-XBP1 and ATF6 target genes are activated in AD and ALS, which may be relevant to the development of new therapeutic strategies. Graphical abstract The endoplasmic reticulum (ER) plays an important role in maintenance of proteostasis through the unfolded protein response (UPR). Two major UPR signalling pathways are mediated by IRE1α and ATF6. Here, we demonstrate that these pathways activate differential gene sets in human post-mortem tissues derived from amyotrophic lateral sclerosis (ALS) compared to Alzheimer's disease (AD) cases. Our results identify IRE1α and ATF6 specific targets that can have major implications in the development of new therapeutic strategies and potential biomarkers.
Collapse
Affiliation(s)
- L Montibeller
- Neurogenetics Group, Division of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - J de Belleroche
- Neurogenetics Group, Division of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK.
| |
Collapse
|
41
|
Noël A, Zhou L, Foveau B, Sjöström PJ, LeBlanc AC. Differential susceptibility of striatal, hippocampal and cortical neurons to Caspase-6. Cell Death Differ 2018; 25:1319-1335. [PMID: 29352267 PMCID: PMC6030053 DOI: 10.1038/s41418-017-0043-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 10/20/2017] [Accepted: 11/17/2017] [Indexed: 12/31/2022] Open
Abstract
Active cysteinyl protease Caspase-6 is associated with early Alzheimer and Huntington diseases. Higher entorhinal cortex and hippocampal Caspase-6 levels correlate with lower cognitive performance in aged humans. Caspase-6 induces axonal degeneration in human primary neuron cultures and causes inflammation and neurodegeneration in mouse hippocampus, and age-dependent memory impairment. To assess whether Caspase-6 causes damage to another neuronal system, a transgenic knock-in mouse overexpressing a self-activated form of Caspase-6 five-fold in the striatum, the area affected in Huntington disease, and 2.5-fold in the hippocampus and cortex, was generated. Detection of Tubulin cleaved by Caspase-6 confirmed Caspase-6 activity. The Caspase-6 expressing mice and control littermates were subjected to behavioral tests to assess Huntington disease-relevant psychiatric, motor, and cognitive deficits. Depression was excluded with the forced swim and sucrose consumption tests. Motor deficits were absent in the nesting, clasping, rotarod, vertical pole, gait, and open field analyzes. However, Caspase-6 mice developed age-dependent episodic and spatial memory deficits identified by novel object recognition, Barnes maze and Morris water maze assays. Neuron numbers were maintained in the striatum, hippocampus, and cortex. Microglia and astrocytes were increased in the hippocampal stratum lacunosum molecular and in the cortex, but not in the striatum. Synaptic mRNA profiling identified two differentially expressed genes in transgenic hippocampus, but none in striatum. Caspase-6 impaired synaptic transmission and induced neurodegeneration in hippocampal CA1 neurons, but not in striatal medium spiny neurons. These data revealed that active Caspase-6 in the striatal medium spiny neurons failed to induce inflammation, neurodegeneration or behavioral abnormalities, whereas active Caspase-6 in the cortex and hippocampus impaired episodic and spatial memories, and induced inflammation, neuronal dysfunction, and neurodegeneration. The results indicate age and neuronal subtype-dependent Caspase-6 toxicity and highlight the importance of targeting the correct neuronal subtype to identify underlying molecular mechanisms of neurodegenerative diseases.
Collapse
Affiliation(s)
- Anastasia Noël
- Bloomfield Center for Research in Aging, Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Chemin Côte Ste Catherine, Montreal, QC, H3A 2B4, Canada
- Department of Neurology and Neurosurgery, McGill University, 3755 University Street, Montreal, QC, H3A 2B4, Canada
| | - Libin Zhou
- Bloomfield Center for Research in Aging, Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Chemin Côte Ste Catherine, Montreal, QC, H3A 2B4, Canada
- Department of Neurology and Neurosurgery, McGill University, 3755 University Street, Montreal, QC, H3A 2B4, Canada
- Department of Anatomy and Cell Biology, McGill University, 3755 University Street, Montreal, QC, H3A 2B4, Canada
| | - Bénédicte Foveau
- Bloomfield Center for Research in Aging, Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Chemin Côte Ste Catherine, Montreal, QC, H3A 2B4, Canada
- Department of Neurology and Neurosurgery, McGill University, 3755 University Street, Montreal, QC, H3A 2B4, Canada
| | - P Jesper Sjöström
- Department of Neurology and Neurosurgery, McGill University, 3755 University Street, Montreal, QC, H3A 2B4, Canada
- Centre for Research in Neuroscience, The BRAIN Program, The Research Institute of the McGill University Health Centre, Montreal General Hospital, Montréal, QC, H3G 1A4, Canada
| | - Andréa C LeBlanc
- Bloomfield Center for Research in Aging, Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Chemin Côte Ste Catherine, Montreal, QC, H3A 2B4, Canada.
- Department of Neurology and Neurosurgery, McGill University, 3755 University Street, Montreal, QC, H3A 2B4, Canada.
- Department of Anatomy and Cell Biology, McGill University, 3755 University Street, Montreal, QC, H3A 2B4, Canada.
| |
Collapse
|
42
|
Bhuvanendran S, Kumari Y, Othman I, Shaikh MF. Amelioration of Cognitive Deficit by Embelin in a Scopolamine-Induced Alzheimer's Disease-Like Condition in a Rat Model. Front Pharmacol 2018; 9:665. [PMID: 29988493 PMCID: PMC6026638 DOI: 10.3389/fphar.2018.00665] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 06/04/2018] [Indexed: 12/25/2022] Open
Abstract
Embelin (2,5-dihydroxy-3-undecyl-1,4-benzoquinone) is one of the active components (2.3%) found in Embelia ribes Burm fruits. As determined via in vitro AChE inhibition assay, embelin can inhibit the acetylcholinesterase enzyme. Therefore, embelin can be utilized as a therapeutic compound, after further screening has been conducted for its use in the treatment of Alzheimer's disease (AD). In this study, the nootropic and anti-amnesic effects of embelin on scopolamine-induced amnesia in rats were evaluated. Rats were treated once daily with embelin (0.3 mg/kg, 0.6 mg/kg, 1.2 mg/kg) and donepezil (1 mg/kg) intraperitoneally (i.p.) for 17 days. During the final 9 days of treatment, a daily injection of scopolamine (1 mg/kg) was administered to induce cognitive deficits. Besides that, behavioral analysis was carried out to assess the rats' learning and memory functions. Meanwhile, hippocampal tissues were extracted for gene expression, neurotransmitter, and immunocytochemistry studies. Embelin was found to significantly improve the recognition index and memory retention in the novel object recognition (NOR) and elevated plus maze (EPM) tests, respectively. Furthermore, embelin at certain doses (0.3 mg/kg, 0.6 mg/kg, and 1.2 mg/kg) significantly exhibited a memory-enhancing effect in the absence of scopolamine, besides improving the recognition index when challenged with chronic scopolamine treatment. Moreover, in the EPM test, embelin treated rats (0.6 mg/kg) showed an increase in inflection ratio in nootropic activity. However, the increase was not significant in chronic scopolamine model. In addition, embelin contributed toward the elevated expression of BDNF, CREB1, and scavengers enzymes (SOD1 and CAT) mRNA levels. Next, pretreatment of rats with embelin mitigated scopolamine-induced neurochemical and histological changes in a manner comparable to donepezil. These research findings suggest that embelin is a nootropic compound, which also possesses an anti-amnesic ability that is displayed against scopolamine-induced memory impairment in rats. Hence, embelin could be a promising compound to treat AD.
Collapse
Affiliation(s)
| | - Yatinesh Kumari
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| | | | - Mohd Farooq Shaikh
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| |
Collapse
|
43
|
Acosta D, Powell F, Zhao Y, Raj A. Regional vulnerability in Alzheimer's disease: The role of cell-autonomous and transneuronal processes. Alzheimers Dement 2018; 14:797-810. [PMID: 29306583 PMCID: PMC5994366 DOI: 10.1016/j.jalz.2017.11.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Revised: 11/08/2017] [Accepted: 11/30/2017] [Indexed: 01/21/2023]
Abstract
INTRODUCTION The stereotypical progression of Alzheimer's disease (AD) pathology is not fully understood. The selective impact of AD on distinct regions has led the field to question if innate vulnerability exists. This study aims to determine if the causative factors of regional vulnerability are dependent on cell-autonomous or transneuronal (non-cell autonomous) processes. METHODS Using mathematical and statistical models, we analyzed the contribution of cell-autonomous and non-cell autonomous factors to predictive linear models of AD pathology. RESULTS Results indicate gene expression as a weak contributor to predictive linear models of AD. Instead, the network diffusion model acts as a strong predictor of observed AD atrophy and hypometabolism. DISCUSSION We propose a convenient methodology for identifying genes and their role in determining AD topography, in comparison with network spread. Results reinforce the role of transneuronal network spread on disease progression and suggest that innate gene expression plays a secondary role in seeding and subsequent disease progression.
Collapse
Affiliation(s)
- Diana Acosta
- Department of Neuroscience, Weill Cornell Medical College of Cornell University, New York, NY, USA
| | - Fontasha Powell
- Department of Neuroscience, Weill Cornell Medical College of Cornell University, New York, NY, USA
| | - Yize Zhao
- Department of Healthcare Policy and Research, Weill Cornell Medical College of Cornell University, New York, NY, USA
| | - Ashish Raj
- Department of Neuroscience, Weill Cornell Medical College of Cornell University, New York, NY, USA; Department of Radiology and Biomedical Imaging, UCSF School of Medicine, San Francisco, CA, USA.
| |
Collapse
|
44
|
Jonson M, Nyström S, Sandberg A, Carlback M, Michno W, Hanrieder J, Starkenberg A, Nilsson KPR, Thor S, Hammarström P. Aggregated Aβ1-42 Is Selectively Toxic for Neurons, Whereas Glial Cells Produce Mature Fibrils with Low Toxicity in Drosophila. Cell Chem Biol 2018; 25:595-610.e5. [PMID: 29657084 DOI: 10.1016/j.chembiol.2018.03.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 01/12/2018] [Accepted: 03/12/2018] [Indexed: 10/17/2022]
Abstract
The basis for selective vulnerability of certain cell types for misfolded proteins (MPs) in neurodegenerative diseases is largely unknown. This knowledge is crucial for understanding disease progression in relation to MPs spreading in the CNS. We assessed this issue in Drosophila by cell-specific expression of human Aβ1-42 associated with Alzheimer's disease. Expression of Aβ1-42 in various neurons resulted in concentration-dependent severe neurodegenerative phenotypes, and intraneuronal ring-tangle-like aggregates with immature fibril properties when analyzed by aggregate-specific ligands. Unexpectedly, expression of Aβ1-42 from a pan-glial driver produced a mild phenotype despite massive brain load of Aβ1-42 aggregates, even higher than in the strongest neuronal driver. Glial cells formed more mature fibrous aggregates, morphologically distinct from aggregates found in neurons, and was mainly extracellular. Our findings implicate that Aβ1-42 cytotoxicity is both cell and aggregate morphotype dependent.
Collapse
Affiliation(s)
- Maria Jonson
- Department of Physics, Chemistry and Biology, Linköping University, Linköping SE-581 83, Sweden
| | - Sofie Nyström
- Department of Physics, Chemistry and Biology, Linköping University, Linköping SE-581 83, Sweden
| | - Alexander Sandberg
- Department of Physics, Chemistry and Biology, Linköping University, Linköping SE-581 83, Sweden
| | - Marcus Carlback
- Department of Physics, Chemistry and Biology, Linköping University, Linköping SE-581 83, Sweden
| | - Wojciech Michno
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, 431 80 Mölndal, Sweden
| | - Jörg Hanrieder
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, 431 80 Mölndal, Sweden; Department of Molecular Neuroscience, Institute of Neurology, University College London, London W1C3BG, UK
| | - Annika Starkenberg
- Department of Clinical and Experimental Medicine, Linköping University, Linköping SE-581 85, Sweden
| | - K Peter R Nilsson
- Department of Physics, Chemistry and Biology, Linköping University, Linköping SE-581 83, Sweden
| | - Stefan Thor
- Department of Clinical and Experimental Medicine, Linköping University, Linköping SE-581 85, Sweden
| | - Per Hammarström
- Department of Physics, Chemistry and Biology, Linköping University, Linköping SE-581 83, Sweden.
| |
Collapse
|
45
|
Tau Spreading Mechanisms; Implications for Dysfunctional Tauopathies. Int J Mol Sci 2018; 19:ijms19030645. [PMID: 29495325 PMCID: PMC5877506 DOI: 10.3390/ijms19030645] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 02/20/2018] [Accepted: 02/21/2018] [Indexed: 12/14/2022] Open
Abstract
Tauopathies comprise a group of progressive age-associated neurodegenerative diseases where tau protein deposits are found as the predominant pathological signature (primary tauopathies) or in combination with the presence of other toxic aggregates (secondary tauopathies). In recent years, emerging evidence suggests that abnormal tau accumulation is mediated through spreading of seeds of the protein from cell to cell, favouring the hypothesis of a prion-like transmission of tau to explain the propagation of the pathology. This would also support the concept that the pathology initiates in a very small part of the brain before becoming symptomatic and spreads across the brain over time. To date, many key questions still remain unclear, such as the nature of the tau species involved in the spreading, the precise seeding/template and uptaking mechanisms or the selectivity explaining why certain neurons are affected and some others are not. A better understanding of the tau spreading machinery will contribute to the development of new therapeutic approaches focused on halting the abnormal propagation, offering also new perspectives for early diagnosis and preventive therapies. In this review, we will cover the most recent advances in tau spreading mechanisms as well as the implications of these findings for dysfunctional tauopathies.
Collapse
|
46
|
What Is Our Current Understanding of PrP Sc-Associated Neurotoxicity and Its Molecular Underpinnings? Pathogens 2017; 6:pathogens6040063. [PMID: 29194372 PMCID: PMC5750587 DOI: 10.3390/pathogens6040063] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 11/21/2017] [Accepted: 11/27/2017] [Indexed: 01/15/2023] Open
Abstract
The prion diseases are a collection of fatal, transmissible neurodegenerative diseases that cause rapid onset dementia and ultimately death. Uniquely, the infectious agent is a misfolded form of the endogenous cellular prion protein, termed PrPSc. Despite the identity of the molecular agent remaining the same, PrPSc can cause a range of diseases with hereditary, spontaneous or iatrogenic aetiologies. However, the link between PrPSc and toxicity is complex, with subclinical cases of prion disease discovered, and prion neurodegeneration without obvious PrPSc deposition. The toxic mechanisms by which PrPSc causes the extensive neuropathology are still poorly understood, although recent advances are beginning to unravel the molecular underpinnings, including oxidative stress, disruption of proteostasis and induction of the unfolded protein response. This review will discuss the diseases caused by PrPSc toxicity, the nature of the toxicity of PrPSc, and our current understanding of the downstream toxic signaling events triggered by the presence of PrPSc.
Collapse
|
47
|
Alibhai J, Diack A, Manson J. Disease-associated protein seeding suggests a dissociation between misfolded protein accumulation and neurodegeneration in prion disease. Prion 2017; 11:381-387. [PMID: 29023184 PMCID: PMC5786362 DOI: 10.1080/19336896.2017.1378289] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Chronic neurodegenerative diseases, such as prion diseases or Alzheimer's disease, are associated with progressive accumulation of host proteins which misfold and aggregate. Neurodegeneration is restricted to specific neuronal populations which show clear accumulation of misfolded proteins, whilst neighbouring neurons remain unaffected. Such data raise interesting questions about the vulnerability of specific neuronal populations to neurodegeneration and much research has concentrated only on the mechanisms of neurodegeneration in afflicted neuronal populations. An alternative, undervalued and almost completely unstudied question however is how and why neuronal populations are resilient to neurodegeneration. One potential answer is unaffected regions do not accumulate misfolded proteins, thus mechanisms of neurodegeneration do not become activated. In this perspectives, we discuss novel data from our laboratories which demonstrate that misfolded proteins do accumulate in regions of the brain which do not show evidence of neurodegeneration and further evidence that microglial responses may define the severity of neurodegeneration.
Collapse
Affiliation(s)
- James Alibhai
- The National CJD Research and Surveillance Unit, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Abigail Diack
- Neurobiology Division, The Roslin Institute, University of Edinburgh, Easter Bush, UK
| | - Jean Manson
- Neurobiology Division, The Roslin Institute, University of Edinburgh, Easter Bush, UK,Correspondence to: Jean Manson; The Roslin Institute, University of Edinburgh, Easter Bush, UK, EH25 9RG;
| |
Collapse
|
48
|
Mezias C, LoCastro E, Xia C, Raj A. Connectivity, not region-intrinsic properties, predicts regional vulnerability to progressive tau pathology in mouse models of disease. Acta Neuropathol Commun 2017; 5:61. [PMID: 28807028 PMCID: PMC5556602 DOI: 10.1186/s40478-017-0459-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Accepted: 07/15/2017] [Indexed: 12/17/2022] Open
Abstract
Spatiotemporal tau pathology progression is regarded as highly stereotyped within each type of degenerative condition. For instance, AD has a progression of tau pathology consistently beginning in the entorhinal cortex, the locus coeruleus, and other nearby noradrenergic brainstem nuclei, before spreading to the rest of the limbic system as well as the cingulate and retrosplenial cortices. Proposed explanations for the consistent spatial patterns of tau pathology progression, as well as for why certain regions are selectively vulnerable to exhibiting pathology over the course of disease generally focus on transsynaptic spread proceeding via the brain's anatomic connectivity network in a cell-independent manner or on cell-intrinsic properties that might render some cell populations or regions uniquely vulnerable. We test connectivity based explanations of spatiotemporal tau pathology progression and regional vulnerability against cell-intrinsic explanation, using regional gene expression profiles as a proxy. We find that across both exogenously seeded and non-seeded tauopathic mouse models, the connectivity network provides a better explanation than regional gene expression profiles, even when such profiles are limited to specific sets of tau risk-related genes only. Our results suggest that, regardless of the location of pathology initiation, tau pathology progression is well characterized by a model positing entirely cell-type and molecular environment independent transsynaptic spread via the mouse brain's connectivity network. These results further suggest that regional vulnerability to tau pathology is mainly governed by connectivity with regions already exhibiting pathology, rather than by cell-intrinsic factors.
Collapse
Affiliation(s)
- Chris Mezias
- Department of Neuroscience, Weill Cornell Medicine of Cornell University, New York, USA.
| | - Eve LoCastro
- Department of Radiology, Weill Cornell Medicine of Cornell University, New York, USA
| | - Chuying Xia
- Department of Neuroscience, Weill Cornell Medicine of Cornell University, New York, USA
| | - Ashish Raj
- Department of Neuroscience, Weill Cornell Medicine of Cornell University, New York, USA.
- Department of Radiology, Weill Cornell Medicine of Cornell University, New York, USA.
| |
Collapse
|
49
|
Bistaffa E, Rossi M, De Luca CMG, Moda F. Biosafety of Prions. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 150:455-485. [PMID: 28838674 DOI: 10.1016/bs.pmbts.2017.06.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Prions are the infectious agents that cause devastating and untreatable disorders known as Transmissible Spongiform Encephalopathies (TSEs). The pathologic events and the infectious nature of these transmissible agents are not completely understood yet. Due to the difficulties in inactivating prions, working with them requires specific recommendations and precautions. Moreover, with the advent of innovative technologies, such as the Protein Misfolding Cyclic Amplification (PMCA) and the Real Time Quaking-Induced Conversion (RT-QuIC), prions could be amplified in vitro and the infectious features of the amplified products need to be carefully assessed.
Collapse
Affiliation(s)
- Edoardo Bistaffa
- IRCCS Foundation Carlo Besta Neurological Institute, Milan, Italy; Scuola Internazionale Superiore di Studi Avanzati (SISSA), Trieste, Italy
| | - Martina Rossi
- Scuola Internazionale Superiore di Studi Avanzati (SISSA), Trieste, Italy
| | - Chiara M G De Luca
- IRCCS Foundation Carlo Besta Neurological Institute, Milan, Italy; Università degli Studi di Pavia, Pavia, Italy
| | - Fabio Moda
- IRCCS Foundation Carlo Besta Neurological Institute, Milan, Italy.
| |
Collapse
|
50
|
Iaccarino L, Moresco RM, Presotto L, Bugiani O, Iannaccone S, Giaccone G, Tagliavini F, Perani D. An In Vivo 11C-(R)-PK11195 PET and In Vitro Pathology Study of Microglia Activation in Creutzfeldt-Jakob Disease. Mol Neurobiol 2017; 55:2856-2868. [PMID: 28455699 DOI: 10.1007/s12035-017-0522-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 04/06/2017] [Indexed: 01/08/2023]
Abstract
Microgliosis is part of the immunobiology of Creutzfeldt-Jakob disease (CJD). This is the first report using 11C-(R)-PK11195 PET imaging in vivo to measure 18 kDa translocator protein (TSPO) expression, indexing microglia activation, in symptomatic CJD patients, followed by a postmortem neuropathology comparison. One genetic CJD (gCJD) patient, two sporadic CJD (sCJD) patients, one variant CJD (vCJD) patient (mean ± SD age, 47.50 ± 15.95 years), and nine healthy controls (mean ± SD age, 44.00 ± 11.10 years) were included in the study. TSPO binding potentials were estimated using clustering and parametric analyses of reference regions. Statistical comparisons were run at the regional and at the voxel-wise levels. Postmortem evaluation measured scrapie prion protein (PrPSc) immunoreactivity, neuronal loss, spongiosis, astrogliosis, and microgliosis. 11C-(R)-PK11195-PET showed a significant TSPO overexpression at the cortical level in the two sCJD patients, as well as thalamic and cerebellar involvement; very limited parieto-occipital activation in the gCJD case; and significant increases at the subcortical level in the thalamus, basal ganglia, and midbrain and in the cerebellum in the vCJD brain. Along with misfolded prion deposits, neuropathology in all patients revealed neuronal loss, spongiosis and astrogliosis, and a diffuse cerebral and cerebellar microgliosis which was particularly dense in thalamic and basal ganglia structures in the vCJD brain. These findings confirm significant microgliosis in CJD, which was variably modulated in vivo and more diffuse at postmortem evaluation. Thus, TSPO overexpression in microglia activation, topography, and extent can vary in CJD subtypes, as shown in vivo, possibly related to the response to fast apoptotic processes, but reaches a large amount at the final disease course.
Collapse
Affiliation(s)
- Leonardo Iaccarino
- Vita-Salute San Raffaele University, Via Olgettina 58, 20132, Milan, Italy.,In Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Via Olgettina 58, 20132, Milan, Italy
| | - Rosa Maria Moresco
- Nuclear Medicine Unit, IRCCS San Raffaele Hospital, Via Olgettina 60, 20132, Milan, Italy.,IBFM-CNR, Via F.lli Cervi 93, Segrate, 20090, Milan, Italy.,Department of Health Sciences, University of Milan Bicocca, Piazza dell'Ateneo Nuovo, 1, 20126, Milan, Italy
| | - Luca Presotto
- In Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Via Olgettina 58, 20132, Milan, Italy.,Nuclear Medicine Unit, IRCCS San Raffaele Hospital, Via Olgettina 60, 20132, Milan, Italy
| | - Orso Bugiani
- IRCCS Foundation "Carlo Besta" Neurological Institute, Via Celoria 11, 20133, Milan, Italy
| | - Sandro Iannaccone
- Neurological Rehabilitation Unit, Clinical Neurosciences Department, IRCCS San Raffaele Hospital, Via Olgettina 60, 20132, Milan, Italy
| | - Giorgio Giaccone
- IRCCS Foundation "Carlo Besta" Neurological Institute, Via Celoria 11, 20133, Milan, Italy
| | - Fabrizio Tagliavini
- IRCCS Foundation "Carlo Besta" Neurological Institute, Via Celoria 11, 20133, Milan, Italy
| | - Daniela Perani
- Vita-Salute San Raffaele University, Via Olgettina 58, 20132, Milan, Italy. .,In Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Via Olgettina 58, 20132, Milan, Italy. .,Nuclear Medicine Unit, IRCCS San Raffaele Hospital, Via Olgettina 60, 20132, Milan, Italy.
| |
Collapse
|